69
Views
1
CrossRef citations to date
0
Altmetric
Review

A review of pioglitazone HCL and glimepiride in the treatment of type 2 diabetes

&
Pages 721-731 | Published online: 28 Dec 2022

Abstract

Type 2 diabetes (T2D) is a progressive disorder with a consistent and steady increase in glycosylated hemoglobin (HbA1c) over time associated with enhanced risk of micro- and macrovascular complications and a substantial reduction in life expectancy. There are three major pathophysiologic abnormalities associated with T2D: impaired insulin secretion, excessive hepatic glucose output, and insulin resistance in skeletal muscle, liver, and adipose tissue. These defects have been treated in clinical praxis by use of oral insulin secretagogues (sulfonylureas/glinides) or insulin, biguanides, and thiazolidinediones (TZDs) respectively. Pioglitazone HCL is an insulin sensitizer in the TZD family and glimepiride is an insulin secretagogue in the SU family. This article reviews mechanisms of action and clinical data behind the use of these two commonly used oral hypoglycemic agents with documented efficacy and good safety profile of once-daily administration, alone or in combination with insulin or metformin, in the management of T2D in terms of glycemic and non-glycemic effects, tolerability and side effects, and impact on vascular health.

Search criteria

Studies were obtained from searches of databases MEDLINE, Cochrane Database of Systematic Reviews, and Cochrane Central Register of Controlled Trials, using search terms; type 2 diabetes, pioglitazone, glimepiride, sulfonylureas, and thiazolidinediones limiting for English language. The last search was conducted in March 2007.

Mechanisms of action

Thiazolidinediones

Thiazolidinediones (TZDs) are agonists of peroxisome-proliferator activated receptorsγ(PPARγ)(CitationLehmann et al 1995; CitationHenry 1997), which are nuclear receptors, composed of three major isoforms (α, β/δ, and γ). PPARγ is a transcription factor, when activated by TZDs, promoting transcription of insulin-sensitive genes involved in fatty acid and glucose uptake and lipogenesis and thereby enhance or partially mimic selective actions of insulin (CitationSaltiel and Olefsky 1996). PPARγ is also essential for normal adipocyte differentiation and proliferation (CitationLemberger et al 1996). PPARγ is expressed in key target tissues for insulin action, most abundantly in adipose tissue but also in skeletal muscle, liver, pancreatic β-cells, vascular endothelium, and macrophages (CitationDubois et al 2000; CitationWillson et al 2001). The glucose-lowering effect of TZDs is attributed to increased peripheral glucose disposal (CitationNolan et al 1994; CitationMiyazaki et al 2001a, Citationb; CitationMiyazaki et al 2002a) and decreased hepatic glucose output (CitationMiyazaki et al 2001a). There is also evidence of increased insulin secretory response in subjects with type 2 diabetes (T2D) following treatment with TZDs (CitationMiyazaki et al 2002b; CitationWallace et al 2004), and a recent in vitro study of acute effects of the PPARγ agonist pioglitazone on human islets from subjects both without and with T2D suggests that short-term pretreatment with pioglitazone primes both healthy and diabetic human islets for enhanced glucose-sensitive insulin secretion (CitationZhang et al 2006). TZDs have been shown to cause redistribution of fat from hepatic and visceral adipose tissue to subcutaneous adipose tissue (CitationMiyazaki et al 2002a; CitationShadid and Jensen 2003; CitationRasouli et al 2005; CitationSmith et al 2005), and in this way possibly saving tissues such as liver, skeletal muscle, and pancreatic β-cells from lipotoxicity.

Two TZDs are currently approved for treatment of hyperglycemia in T2D, rosiglitazone and pioglitazone, which are both potent and highly selective agonists for PPARγ, being very similar in mechanism of action, effect on hyperglycemia, and side effect profiles. It is important to keep in mind that these drugs need insulin as a co-factor for PPAR agonism (CitationFeinglos and Bethel 1998), and if used at late stages of T2D, when insulinopenia has already developed, the full effect cannot be expected.

Pioglitazone HCL is a PPARγ agonist that increases both insulin-stimulated glucose uptake in peripheral tissues (CitationMiyazaki et al 2001b) and insulin sensitivity in hepatic and adipose tissue (CitationKawamori et al 1998; CitationMiyazaki et al 2004), thereby lowering plasma glucose both as single agent and in combination with other oral hypoglycemic agents (OHA) and/or insulin (CitationAronoff et al 2000; CitationRosenblatt et al 2001; CitationScherbaum and Goke 2002; CitationHerz et al 2003). Pioglitazone has also been shown to have multiple beneficial effects on lipid metabolism (CitationWinkler et al 2003; CitationKhan et al 2004; CitationPerez et al 2004; CitationBetteridge and Verges 2005; CitationGoldberg et al 2005), endothelial function (CitationForst et al 2005b; CitationMartens et al 2005), atherogenesis (CitationLangenfeld et al 2005; CitationMazzone et al 2006), fibrinolysis (CitationDerosa et al 2005b), and immune function (CitationMarx et al 2000; CitationGosset et al 2001; CitationMartens et al 2006).

Pioglitazone has in addition shown some minor activation of PPARα (CitationForman et al 1995; CitationLehmann et al 1995). This activation has been linked to lowering of plasma triglyceride (TG) levels. PPARα activation is also known to have anti-inflammatory effects and preventive effect on arteriosclerosis (CitationRubins et al 1999; CitationDuez et al 2002).

Sulfonylureas

Sulfonylureas (SUs) are widely used drugs in the treatment of T2D. They exert their insulin-releasing effect mainly by inhibiting ATP-sensitive potassium channels. In the pancreatic β-cell this action induces depolarization of the cell membrane, allowing an influx of calcium in the cell. This in turn induces insulin release into the blood (CitationAshcroft and Rorsman 1989). The insulin release is not modified by plasma glucose and the SU action can thus induce hypoglycemia.

It is, however, also evident that SUs have insulin-releasing effects independent of ATP-sensitive potassium channels. Tolbutamide, glibenclamide, and glipizide were shown to increase insulin secretion in human β-cells also when the membrane of the β-cell was voltage-clamped and thus Ca-flux was unaffected (CitationEliasson et al 1996). It was also suggested that this action was mediated by protein kinase C (PKC), since the stimulatory action of tolbutamide was abolished by a PKC inhibitor. Recent studies of rat β-cells have shown that glibenclamide inhibits activity of carnitine palmitoyltransferase 1 (CPT 1) which in turn switches fatty acid catabolism to synthesis of PKC-activating lipids and CPT 1 may thus play a major role in SU-mediated insulin release (CitationLehtihet et al 2003). In this context it is also interesting to note that it was shown long ago (CitationHellman et al 1984) that glibenclamide accumulates in the β-cell during chronic treatment.

ATP-sensitive potassium channels are not specific to the pancreatic β-cell but found in a variety of cells throughout the human organism. These cells include vascular smooth muscle cells (CitationQuayle et al 1997) vascular endothelium, cardiac myocytes (CitationNichols and Lederer 1991) and some brain neurons (CitationYamada et al 2001). ATP-sensitive potassium channels are also found on the inner membranes of mitochondria (CitationDahlem et al 2004). In all mentioned cell types, opening of KATP channels in response to metabolic stress leads to inhibition of electrical activity and are in that way involved in the response of both cardiac and cerebral ischemia. The K channels consist of two entities: Kir6 (belonging to the family of inwardly rectifying K+ (Kir) channels) and a SU receptor, SUR. There is more than one isoform of both Kir6 (Kir6.1, Kir6.2) and SUR (SUR1, SUR2A, SUR2B) (CitationProks et al 2002). Kir6 and SUR are associated in different ways in body tissues and show different sensitivity to various K blockers. It is well established that SU can block ATP-sensitive potassium channels also in cells outside the pancreas. It has been proposed that ischemic preconditioning is a mechanism of protection against myocardial ischemia. It has been described as an adaptive response of the myocardium to repeated episodes of ischemia and the proposed mechanism is an opening of ATP-sensitive potassium channels in the mitochondria. The blocking of potassium channels in the heart by SU medication has given rise to a long-standing concern regarding the potential adverse effects of SU medication in the event of ischemic heart disease. See below for a review of existing data.

Clinical trials

Pioglitazone HCL in treatment of T2D

Effect on plasma glucose, insulin sensitivity and lipids

Pioglitazone 7.5–45 mg daily has been compared with placebo in several randomized, double blind, parallel-group studies in patients with T2D. Pioglitazone 15, 30, and 45 mg produced significant improvements in HbA1c and fasting plasma glucose (FPG). Mean change in HbA1c from baseline ranged from −0.3% to −1.8% and up to −2.6% when adjusted for difference from placebo. Pioglitazone also produced significant decreases in TG and significant increases in high density lipoprotein (HDL) (CitationAronoff et al 2000; CitationRosenblatt et al 2001; CitationScherbaum and Goke 2002; CitationHerz et al 2003).

In comparative/combination trials with metformin, in a 1-year, active-comparator trial, patients poorly controlled with SU were randomly assigned to metformin or pioglitazone. Both treatments resulted in comparable reductions in HbA1c and FPG with sustained glycemic control throughout the study (CitationHanefeld et al 2004). In a combination trial metformin plus pioglitazone 30 mg was compared with metformin plus placebo in patients with poorly controlled T2D. Treatment with pioglitazone 30 mg plus metformin resulted in significant improvement in HbA1c and FPG compared with placebo plus metformin. In addition, patients treated with metformin plus pioglitazone had a significant reduction in TG and a significant increase in HDL-C compared with metformin plus placebo (CitationEinhorn et al 2000). During two active-comparator trials, treatment with metformin and pioglitazone resulted in equal, significant reduction in HbA1c while insulin sensitivity measured by homeostasis model assessment showed significant improvement in the pioglitazone, but not the metformin group at 32 weeks (CitationPavo et al 2003; CitationSchernthaner et al 2004b).

In comparative/combination trials with SUs, pioglitazone produced comparable reduction in HbA1c and FPG as SU. Pioglitazone produced a slower but sustained reduction in HbA1c compared with an earlier effect in the SU treated group. Insulin sensitivity measured by homeostasis model assessment showed significantly greater improvement during pioglitazone treatment (CitationTan et al 2004a, Citationb). In combination trials, pioglitazone produced significant additional improvement in glycemic control, as measured by FPG and HbA1c. Secondary efficacy measures, including TG and HDL, also showed significant improvement after adding pioglitazone to SU (CitationKipnes et al 2001; CitationHanefeld et al 2004).

Improvement in HbA1c was sustained throughout the double-blind phase and for 72 and 104 weeks in 2 open-label extension trials when combining pioglitazone with metformin or glibenclamide (CitationEinhorn et al 2000; CitationTan et al 2004b).

Both pioglitazone and rosiglitazone are also approved for triple combination with metformin and a SU, producing significant additional decrease in HbA1c and seem to be equally effective (CitationBalfour and Plosker 1999; CitationGillies and Dunn 2000; CitationTran et al 2006).

In combination/comparative trials of pioglitazone and insulin, pioglitazone, when added to stable insulin regimens, significantly improved HbA1c and FPG in patients with T2D. Pioglitazone treatment also provided significant benefit with respect to plasma HDL and TG levels and was as effective as a single injection of NPH at bedtime (CitationRosenstock et al 2002; CitationAljabri et al 2004; CitationMattoo et al 2005; CitationDavidson et al 2006).

Side effects

During clinical trials, adverse events have been mild to moderate in severity with an overall frequency of adverse events being similar across the treatment groups (pioglitazone vs metformin, SU, or insulin). Adverse event profiles were different between treatment groups. During treatment with pioglitazone, edema and weight gain were reported more frequently, whereas gastrointestinal adverse events were reported more frequently in the metformin group. Hypoglycemia occurred at a higher incidence in patients treated with a SU or insulin (CitationAljabri et al 2004; CitationSchernthaner et al 2004b; CitationCharbonnel et al 2005; CitationBelcher et al 2005; CitationJain et al 2006). There were no signs of hepatotoxicity in these studies. On the contrary there was a decrease in liver enzymes which also has been shown in other studies (CitationDorkhan et al 2006) and in the DREAM study with rosiglitazone (CitationGerstein et al 2006). Interestingly, several pilot trials have consistently shown positive effects of TZDs in the treatment of nonalcoholic steatohepatitis (NASH) (CitationCaldwell et al 2001; CitationNeuschwander-Tetri et al 2003; CitationSanyal et al 2004; CitationTiikkainen et al 2004) and Belfort et al reported recently in a placebo-controlled trial that administration of pioglitazone led to metabolic and histologic improvement in subjects with NASH (CitationBelfort et al 2006). However, an increase in incidence of heart failure (CitationDormandy et al 2005) and bone loss in older women (CitationKahn et al 2006; CitationSchwartz et al 2006) are the greatest matters of concern with TZDs at present, warranting further investigation.

Potential cardiovascular effects of pioglitazone HCL

As monotherapy or in combination with SU, metformin, or insulin, pioglitazone has shown consistent improvements in TG and HDL levels with no significant effect on total or LDL cholesterol, the greatest degree of improvement occurring at the highest dosage in most cases. These changes ranged from about a 16% decrease in TG to a 12%–19% increase in HDL (CitationAronoff et al 2000; CitationRosenblatt et al 2001; CitationScherbaum and Goke 2002; CitationHerz et al 2003). Treatment with pioglitazone seems to be associated with greater beneficial effects on blood lipid levels than treatment with rosiglitazone (CitationBoyle et al 2002; CitationGoldberg et al 2005). Pioglitazone also has shown to reduce dense LDL significantly and independently from fasting triglycerides and HDL cholesterol, suggesting the antiatherogenic potential of pioglitazone may be greater than that expected from its effects on triglycerides, LDL, and HDL cholesterol alone (CitationWinkler et al 2003). In a study by Berhanu et al, there was marked improvements in lipid profiles in terms of LDL particle size and apolipoproteins after treatment conversion from rosiglitazone to pioglitazone in patients with T2D (CitationBerhanu et al 2006).

There is some evidence on beneficial effect of TZDs on hypertension in patients with (CitationFullert et al 2002; CitationGerber et al 2003; CitationFukuen et al 2005; CitationDerosa et al 2005a; CitationIRIS 2007; CitationDormandy et al 2005) and without T2D (CitationFullert et al 2002).

TZDs have also been shown to increase adiponectin levels, which are reduced in obesity and negatively correlated to insulin resistance and development of arteriosclerosis (CitationForst et al 2005a; CitationPfutzner et al 2005a; CitationDorkhan et al 2006). However, the mechanism is not well understood yet but a recent study proposed that increased adiponectin production by pioglitazone was mediated through suppression of SOCS3 expression, playing an important role in improved insulin sensitivity (CitationKanatani et al 2007).

In other studies there have been evidences of reduction, regression or slowing progression of carotid intima media thickness (CIMT) (CitationNakamura et al 2004; CitationLangenfeld et al 2005; CitationMazzone et al 2006) in patients with T2D when treated with pioglitazone as well as reduced neointimal tissue proliferation after coronary stent implantation and reduced in-stent restenosis (CitationTakagi et al 2003; CitationMarx et al 2005; CitationNishio et al 2006). The study by Mazzone et al was a randomized, double-blind, comparator-controlled, multicenter trial including 462 adults with T2D, comparing effect of pioglitazone vs glimepiride on CIMT during 18 months of treatment. The change in CIMT was less with pioglitazone vs glimepiride, and pioglitazone slowed progression of maximum CIMT compared with glimepiride similarly across prespecified subgroups based on age, sex, systolic blood pressure, duration of diabetes, body mass index, HbA1c value, and statin use. In a pioneer study there was specific evidence of anti-inflammatory and antiatherogenic effect of pioglitazone versus glimepiride in terms of reduction in high-sensitiveity C-reactive protein, matrix metalloproteinase, and monocyte chemoattractant protein, independent of glycemic control (CitationPfutzner et al 2005b).

Data from several animal and human studies support the notion that TZDs even reduce urine albumin excretion and may prevent development of renal injury. There is, however, a relative lack of evidence demonstrating the effects of TZDs on hard renal outcomes as reviewed by Sarafidis and Bakris (2006) and there is need for studies with the particular objective of possible renoprotective effect of TZDs.

The PROspective pioglitAzone Clinical Trial In macroVascular Events (PROactive) study was the first randomized, double-blind outcome study in patients with T2D managed with diet and/or oral blood glucose-lowering drugs and/or insulin who had a history of macrovascular disease, assessing the effect of pioglitazone on the secondary prevention of macrovascular events. A total of 5238 patients were randomized with the cohort of patients, a typical type 2 diabetic population at high risk of further macrovascular events. The results were presented in 2005 after an average time of observation of 34.5 months. Treatment with pioglitazone reduced the secondary endpoint of combined all-cause mortality, non-fatal myocardial infarction, and stroke by 16% (CitationDormandy et al 2005), at the expense of increased heart failure (CitationYki-Jarvinen 2005). A recently published subgroup analysis from PROactive demonstrated that pioglitazone reduced the risk of recurrent stroke significantly in high-risk patients with T2D (CitationWilcox et al 2007).

Glimepiride in treatment of T2D

Effect on plasma glucose

Two systematic reviews on the effects and side effects of glimepiride (CitationCampbell 1998; CitationInzucchi 2002) have been helpful in identifying randomized controlled trials (RCTs). CitationDraeger et al (1996) found equivalent reductions in HbA1c after 12 months’(tm) treatment with glimepiride (1–8 mg) or glibenclamide (2.5–20 mg) after titration to a common target. End mean HbA1c in the glimepiride group was 8.4% and in the glibenclamide group 8.3%. The study was conducted in 1044 patients with T2D aged 26–81 years. All patients were previously treated with glibenclamide for ≥2 months; half the study population was thus transferred to treatment with glimepiride and half remained on glibenclamide. The purpose of the study was to show non-inferiority of glimepiride vs glibenclamide. Another RCT (CitationDills and Schneider 1996) studied 577 patients, mean age 59.5 years, with type 2 diabetes previously treated with diet or any SU. No significant difference in HbA1c was found at 52 weeks after treatment with glimepiride 1–12 mg or glibenclamide 1.25–15.00 mg titrated to pre-determined targets. HbA1c was reduced from 8.50 ± 1.20% (mean ± SD) at baseline to 8.24 ± 1.51% for glimepiride and from 8.50 ± 1.30% at baseline to 8.28 ± 1.48% for glibenclamide. Also in this study the main objective was to show non-inferiority of glimepiride compared with glibenclamide.

One study was found (CitationSchernthaner et al 2004a) that compared adding modified release gliclazide (gliclazide MR) 30–120 mg or glimepiride 1–6 mg daily to current treatment (diet alone, metformin, or acarbose) in 845 patients with type 2 diabetes. It found no significant difference in HbA1c at 27 weeks between gliclazide MR and glimepiride. HbA1c decreased similarly in both groups, from 8.4% to 7.2% on gliclazide MR and from 8.2% to 7.2% on glimepiride.

Several RCTs have been compared glimepiride with placebo as therapy added to lifestyle changes in drug-naive patients. In one such study (CitationLuis Bautista et al 2003) glimepiride 1–4 mg (n = 48, mean age 48.4) or placebo (n = 22, mean age 50.7) was added to diet/exercise advice in Mexican Americans. Not surprisingly the HbA1c reduction from baseline to end-point was statistically significant in favor of glimepiride after 14 weeks.

Side effects

CitationRosenstock et al (1996) reported no episodes of hypoglycemia (p-glucose <3.3 mmol/L) with either glimepiride 8–16 mg/day or placebo during a 14-week study in 416 patients previously treated with any drug in monotherapy. CitationLuis Bautista et al (2003) found no significant difference in self-assessment of hypoglycemia reported over 14 weeks between patients taking glimepiride 1–4 mg or placebo and no confirmed episodes of hypoglycemia (plasma glucose <3.1 mmol/L). The weight gain was significantly higher in the glimepiride group compared with the placebo group.

CitationDraeger et al (1996) found a lower incidence of hypoglycemia in people treated with glimepiride 1–8 mg compared with glibenclamide 2.5–20 mg. The incidence was 60 episodes with glimepiride and 74 episodes with glibenclamide. The effect on HbA1c was the same in both groups, as reported above. The study by CitationDills and Schneider (1996) found a lower incidence of symptomatic hypoglycemia (12% and 17% respectively) in people treated with glimepiride 1–12 mg compared with glibenclamide 1.25–15.00 mg over 12 months.

Potential cardiovascular effects of SUs in general and glimepiride in particular

Initial concern for this issue was raised in the early 1970s when the University Group Diabetes Program assessed the efficacy of oral hypoglycemic treatment compared with insulin and diet alone in the prevention of cardiovascular complications. They demonstrated a significantly higher cardiovascular mortality in patients treated with SUs compared with diet alone (CitationProut 1971; CitationKaram et al 1975). The United Kingdom Prospective Diabetes Study (UKPDS) attempted to answer the question of whether improved glycemic control reduced the risk of cardiovascular death in patients who were taking insulin or SUs (CitationUKPDS 1998a, Citationb). In that study no detrimental effect of SUs was noted, and the UKPDS is often cited as evidence that SUs do not increase cardiovascular risk to patients with type 2 diabetes. The UKPDS, however, did not ascertain the effect these agents had on patients with type 2 diabetes in the setting of acute coronary syndromes, ie, in patients directly at risk of myocardial infarction. The UKPDS patients were also newly diagnosed and known cardiovascular disease was an exclusion criterion; the absolute numbers of cardiovascular events were thus low. In a UKPDS substudy metformin in monotherapy was found to have a beneficial impact on cardiovascular events; this effect was not evident when SU was used in combination with metformin.

One review (CitationProks et al 2002) suggests that the SUR1 subunit of the KATP channel is mainly found in the beta-cell and that the SUR2 subunit of the KATP channel is found in smooth muscle and cardiac cells. It is further proposed that in humans, tolbutamide and gliclazide block channels containing SUR1 whereas glibenclamide, glimepiride, and repaglinide block both types of channels. It is, however, also clear that the inhibition of the KATP channel is not complete and that differences appear between drugs. In an extensive review (CitationQuast et al 2004) the authors concluded that in animal models the coronary vasculature of several species respond to glibenclamide with vasoconstriction and a reduction of coronary blood flow and they also found indications in several studies that the vascular KATP channel plays a major role in the autoregulation of coronary blood flow in the animal and human heart in hypoxia. There has been a general lack of knowledge regarding the role of KAPT channels in the myocardium, the general belief being that the high ATP concentrations in the myocyte keep the KAPT channels closed under normal conditions. In one study, however (CitationZingman et al 2002), it was shown that in mice the knockout of Kir6.2 (the ion-conducting subunit of the KAPT channel) resulted in impaired cardiac performance during sympathetic stimulation, pointing to a possible role of KAPT channels in maintaining cardiac cellular homeostasis not only in response to ischemia but also in the adaptive reaction to stress. The role of SUs in this context has not been studied.

Glimepiride binds predominantly to a 65 kDa subunit protein of the KATP complex while other SUs predominantely bind to a 140 kDa protein (CitationKramer et al 1994). The clinical relevance of these findings remains to be elucidated.

One study in perfused hearts in rats where coronary arteries were occluded by means of a snare (CitationMocanu et al 2001) showed that the beneficial effect of ischemic preconditioning was completely abolished by including glibenclamide in the perfusate but unaffected when including glimepiride. Other studies in animals (CitationGeisen et al 1996) have also shown that glimepiride produced less cardiovascular effect than glibenclamide at equivalent blood glucose decrease in rats and dogs. In rabbit ventricular myocytes (CitationSato et al 2006), tolbutamide, gliclazide, and glimepiride had no effect on the mitochondrial KATP channel and did not affect the (cardioprotective) effects of the KATP-opener diazoxide. Glibenclamide on the other hand interfered with the effect of diazoxide and the authors concluded that glibenclamide interferes with the cellular pathways that confer cardioprotection whereas glimepiride, gliclazide, and tolbutamide do not.

Data from studies in humans are scarce, for obvious reasons. In one study (CitationGhosh et al 2001), tissue samples from the right atrium were obtained from patients of 7 different categories: non-diabetics, patients with type 2 diabetes both treated with KATP blockers and diet only, patients with type 1 diabetes, and patients with 3 different degrees of decreased ejection fraction (EF). Five protocols were used in a randomized fashion: 1. Control, incubation in oxygenated buffer; 2. Ischemia alone; 3. Preconditioning + ischemia; 4. Diazoxide + ischemia; 5. Glibenclamide + preconditioning + ischemia. Preconditioning was made by 5-min ischemia/ 5-min reoxygenation before a 90-min ischemia/120-min reoxygenation. Preconditioning prevented the effects of ischemia in all groups except patients with diabetes type 1 and 2 and those with the lowest EF. Diazoxide prevented effects of ischemia in all groups except patients with diabetes type 1 and 2. Glibenclamide abolished protection in non-diabetics and diet-controlled diabetes groups and did not affect diabetes type 1 and diabetes type 2 patients treated with KATP channel blockers.

One study in healthy male volunteers (CitationBijlstra et al 1996) showed that glibenclamide significantly inhibited the diazoxide-induced increase in forearm blood flow compared with placebo whereas glimepiride did not. The findings have been questioned in a later review (CitationRiveline et al 2003).

There are a few important studies that try to study the role of glimepiride and ischemic preconditioning in real-life myocardial ischemia. In a double-blind RCT (CitationKlepzig et al 1999), 45 predominantly male volunteers were divided into 3 groups when performing balloon angioplasty of high grade coronary artery stenosis. Each patient underwent 3 dilatations, and drugs were administered between dilatation 2 and 3. Balloon pressures and time intervals were identical in all 3 dilatations. Three drugs were infused intravenously in a randomized fashion: placebo, 1 mg glimepiride, or 2 mg glibenclamide. The primary variable was the mean ST segment shift during dilatation. Glimepiride and placebo groups showed a statistically significant reduction of mean ST segment shift between dilatation 2 and 3, interpreted as ischemic preconditioning. The glibenclamide group showed no difference in mean ST segment depression between dilatations. Direct comparison of the two drugs showed no statistically significant difference, as the power of the study was low.

One study from Taiwan (CitationLee and Chou 2003) investigated 20 patients with no diabetes and 23 patients with T2D when performing coronary angioplasty. The non-diabetic patients were given 10 mg glibenclamide (n = 6), 2 mg glimepiride (n = 7) or nothing (n = 7) before the angioplasty together with a glucose infusion to prevent hypoglycemia. No hypoglycemias were reported during the study. Patients with diabetes were taking either glimepiride (n = 12) or glibenclamide (n = 11). In both groups mean ST segment shift decreased between the first and the second balloon inflation in the control group and in the group taking glimepiride but not in the glibenclamide group. When nicorandil, an agent specifically activating mitochondrial KATP channels, was given i.v. to patients with diabetes before angioplasty there was no difference in mean ST segment shift in the glimepiride nor glibenclamide groups.

Combination of pioglitazone HCL and glimepiride in treatment of T2D

In vitro studies demonstrate that glimepiride has the potential to induce PPAR gamma activity, thereby improving insulin resistance (CitationFukuen et al 2005; CitationInukai et al 2005).

In a study of long-term effect of TZD on blood pressure in diabetic patients with metabolic syndrome treated with glimepiride there was significant improvement in systolic- and diastolic blood pressure after 12 months suggesting that the addition of a TZD to the glimepiride treatment of patients with T2D and metabolic syndrome is associated with a significant improvement in the long-term blood pressure control, related to a reduction in insulin-resistance (CitationDerosa et al 2005a). In a multicenter, randomized, double-blind, controlled clinical trial of treatment with pioglitazone or rosiglitazone added to glimepiride in patients with T2D and metabolic syndrome, 1 year of treatment with the pioglitazone combination was associated with significantly reduced plasma lipoprotein (a) levels, a known risk factor for cardiovascular disease, compared with the rosiglitazone combination (CitationDerosa et al 2006).

Discussion

Diabetes is a strong, independent risk factor for cardiovascular disease, causing a 2- to 4-fold increase in cardiovascular mortality in patients with T2D (CitationPanzram 1987; CitationStamler et al 1993; CitationUKPDS 1998a, Citationb; CitationStratton et al 2000). The two largest prospective diabetes studies, the UKPDS and the University Group Diabetes Program (UGDP) showed no statistically significant reduction of cardiovascular endpoints through improved glycemic control. Both pioglitazone and glimepiride are well tolerated and effective in achieving metabolic control in treatment of patients with T2D, as monotherapy or in combination. In spite of massive evidence of beneficial effects of pioglitazone on conventional and non-conventional cardiovascular risk factors, the results from the first prospective outcome study (CitationDormandy et al 2005) are still considered controversial and need confirmation. The latest Cochrane review on the subject has considered 22 RCTs including more than 6000 people treated with pioglitazone. They conclude that until new evidence becomes available, the benefit-risk ratio of pioglitazone remains unclear (CitationRichter et al 2006).

There is no doubt that TZDs have contributed to our understanding of the pathophysiology of T2D and its treatment but there are still some unresolved issues about heart failure that require further study before a positive recommendation regarding their effects on cardiovascular risk reduction can be made. In our opinion, it is very important to estimate risk vs benefit in each individual in order to make appropriate therapeutic choices for patients with T2D.

Adequate circulating insulin is a prerequisite for TZD activity (CitationFeinglos and Bethel 1998) and should be accounted for before initiating therapy, otherwise there is a risk that the full effect of TZDs cannot be achieved. For the practising physician it is of great interest to have instruments to identify individuals at risk of developing heart failure or to monitor cardiac function regularly during treatment. Natriuretic peptides can be candidates for both identifying individuals at risk to develop heart failure and to monitor the effect of TZDs on cardiac load, but there are for now no established recommendations addressing this issue (CitationJernberg et al 2004; CitationKarthikeyan and Lip 2007). Some recent studies have been designed to identify factors associated with TZD effect (CitationPhatak and Yin 2006; CitationWilcox et al 2007) but more studies are needed to help draw guidelines for individually adapted therapy choices as well as studies on markers of cardiovascular risk specifically. Several ongoing outcome studies are to be presented in the near future and these will undoubtedly increase our knowledge in this area. All evidence considered, it seems fair to conclude that the positive cardiovascular effects of TZDs (reduced hyperglycemia, improved dyslipidemia, decreased blood pressure, improved endothelial function, reduced central obesity, decreased inflammation, and decreased microalbuminuria) overweigh the negative ones (weight gain and increased heart failure).

The development of a new generation of TZDs, the partial PPARγ agonists with more specific action, may prove efficacious not only in treatment of T2D but also in prevention of atherosclerosis.

Glimepiride is a long-acting SU of recent origin. The glucose-lowering effects are well documented and comparable with those of other long-acting SUs. There is an ongoing controversy regarding SUs and other KAPT channel blockers and the risk of adverse effects on the heart. Since patients with diabetes have a greatly increased risk of developing and dying from cardiovascular disease, it would be appalling if it were proven that a remedy used for centuries in fact worsened the disease! Few would argue that, in the experimental setting, glimepiride can show effects on the heart and vessels that distinguishes it from its most common comparator, glibenclamide. The discussion has almost exclusively revolved around the phenomenon of ischemic preconditioning (IPC), described as a protective action taken by a heart subjected to a short ischemia and thus alleviating the effects of a longer ischemia. The different effects on the heart by the different KATP channel blockers result from their different affinity to SUR1 (mainly β-cell) and SUR2 (mainly heart and vessels) subunits of the SU (140K) receptor. The important question remains to be answered: do these differences matter in our drug-treated T2D patients? This question is clearly very difficult question to answer, since a prospective RCT is almost impossible to perform, but in a prospective study in France (CitationDanchin et al 2005) 487 patients with acute myocardial infarction and diabetes were identified. Among the 215 patients with previous SU treatment, the in-hospital mortality was 10.2% compared with the non-SU (insulin, diet, other drugs) group mortality of 16.9% (p = 0.035).

The UKPDS remains our most important source of prospective data for risk vs treatment of T2D, and according to the UKPDS there is no obvious increase in cardiovascular risk when treating T2D with SUs.

On the other hand, it can be argued that if potential differences in the benefit of glimepiride can be detected in the experimental setting and treatments are comparable in other ways, we may let this influence our clinical decisions. As with the TZDs, we leave this to the discretion and clinical judgment of the reader.

Abbreviations

FPG, fasting plasma glucose; HbA1c, glycosylated hemoglobin; HDL, high density lipoprotein cholesterol; OHA, oral hypoglycemic agents; PPARs, peroxisome-proliferator activated receptors; RCT, randomized controlled trials; SU, sulfonylurea; T2D, type 2 diabetes; TG, triglycerides; TZD, thiazolidinedione.

References

  • AljabriKKozakSEThompsonDMAddition of pioglitazone or bedtime insulin to maximal doses of sulfonylurea and metformin in type 2 diabetes patients with poor glucose control: a prospective, randomized trialAm J Med2004116230514969650
  • AronoffSRosenblattSBraithwaiteSPioglitazone hydrochloride monotherapy improves glycemic control in the treatment of patients with type 2 diabetes: a 6-month randomized placebo-controlled dose-response study. The Pioglitazone 001 Study GroupDiabetes Care20002316051111092281
  • AshcroftFMRorsmanPElectrophysiology of the pancreatic beta-cellProg Biophys Mol Biol198954871432484976
  • BalfourJAPloskerGLRosiglitazoneDrugs19995792130 discussion 931–210400405
  • BelcherGLambertCEdwardsGSafety and tolerability of pioglitazone, metformin, and gliclazide in the treatment of type 2 diabetesDiabetes Res Clin Pract200570536216002175
  • BelfortRHarrisonSABrownKA placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitisN Engl J Med2006355229730717135584
  • BerhanuPKipnesMSKhanMAEffects of pioglitazone on lipid and lipoprotein profiles in patients with type 2 diabetes and dyslipidaemia after treatment conversion from rosiglitazone while continuing stable statin therapyDiab Vasc Dis Res20063394416784180
  • BetteridgeDJVergesBLong-term effects on lipids and lipoproteins of pioglitazone versus gliclazide addition to metformin and pioglitazone versus metformin addition to sulphonylurea in the treatment of type 2 diabetesDiabetologia20054824778116283239
  • BijlstraPJLuttermanJARusselFGInteraction of sulphonylurea derivatives with vascular ATP-sensitive potassium channels in humansDiabetologia1996391083908877293
  • BoylePJKingABOlanskyLEffects of pioglitazone and rosiglitazone on blood lipid levels and glycemic control in patients with type 2 diabetes mellitus: a retrospective review of randomly selected medical recordsClin Ther2002243789611952022
  • CaldwellSHHespenheideEERedickJAA pilot study of a thiazolidinedione, troglitazone,in nonalcoholic steatohepatitisAm J Gastroenterol2001965192511232700
  • CampbellRKGlimepiride: role of a new sulfonylurea in the treatment of type 2 diabetes mellitusAnn Pharmacother1998321044529793597
  • CharbonnelBSchernthanerGBrunettiPLong-term efficacy and tolerability of add-on pioglitazone therapy to failing monotherapy compared with addition of gliclazide or metformin in patients with type 2 diabetesDiabetologia200548109310415889234
  • DahlemYAHornTFBuntinasLThe human mitochondrial KATP channel is modulated by calcium and nitric oxide: a patch-clamp approachBiochim Biophys Acta20041656465615136158
  • DanchinNCharpentierGLedruFRole of previous treatment with sulfonylureas in diabetic patients with acute myocardial infarction: results from a nationwide French registryDiabetes Metab Res Rev200521143915386810
  • DavidsonJAPerezAZhangJAddition of pioglitazone to stable insulin therapy in patients with poorly controlled type 2 diabetes: results of a double-blind, multicentre, randomized studyDiabetes Obes Metab200681647416448520
  • DerosaGCiceroAFD’AngeloAEffects of 1 year of treatment with pioglitazone or rosiglitazone added to glimepiride on lipoprotein a. and homocysteine concentrations in patients with type 2 diabetes mellitus and metabolic syndrome: a multicenter, randomized, double-blind, controlled clinical trialClin Ther2006286798816861090
  • DerosaGCiceroAFDangeloAThiazolidinedione effects on blood pressure in diabetic patients with metabolic syndrome treated with glimepirideHypertens Res2005a289172416555581
  • DerosaGCiceroAFGaddiAA comparison of the effects of pioglitazone and rosiglitazone combined with glimepiride on prothrombotic state in type 2 diabetic patients with the metabolic syndromeDiabetes Res Clin Pract2005b6951315955382
  • DillsDGSchneiderJClinical evaluation of glimepiride versus glyburide in NIDDM in a double-blind comparative study. Glimepiride/Glyburide Research GroupHorm Metab Res19962842698911977
  • DorkhanMMagnussonMFridAGlycaemic and nonglycaemic effects of pioglitazone in triple oral therapy of patients with type 2 diabetesJ Intern Med20062601253316882276
  • DormandyJACharbonnelBEcklandDJSecondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study PROspective pioglitAzone Clinical Trial In macroVascular Events: a randomised controlled trialLancet200536612798916214598
  • DraegerKEWernicke-PantenKLompHJLong-term treatment of type 2 diabetic patients with the new oral antidiabetic agent glimepiride Amaryl: a double-blind comparison with glibenclamideHorm Metab Res199628419258911976
  • DuboisMPattouFKerr-ConteJExpression of peroxisome proliferator-activated receptor gamma PPARgamma. in normal human pancreatic islet cellsDiabetologia2000431165911043863
  • DuezHChaoYSHernandezMReduction of atherosclerosis by the peroxisome proliferator-activated receptor alpha agonist fenofibrate in miceJ Biol Chem200227748051712377786
  • EinhornDRendellMRosenzweigJPioglitazone hydrochloride in combination with metformin in the treatment of type 2 diabetes mellitus: a randomized, placebo-controlled study. The Pioglitazone 027 Study GroupClin Ther200022139540911192132
  • EliassonLRenstromEAmmalaCPKC-dependent stimulation of exocytosis by sulfonylureas in pancreatic beta cellsScience199627181358628999
  • FeinglosMNBethelMATreatment of type 2 diabetes mellitusMed Clin North Am199882757909706120
  • FormanBMTontonozPChenJ15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gammaCell199583803128521497
  • ForstTHohbergCFuellertSDPharmacological PPARgamma stimulation in contrast to beta cell stimulation results in an improvement in adiponectin and proinsulin intact levels and reduces intima media thickness in patients with type 2 diabetesHorm Metab Res2005a37521716138267
  • ForstTLubbenGHohbergCInfluence of glucose control and improvement of insulin resistance on microvascular blood flow and endothelial function in patients with diabetes mellitus type 2Microcirculation2005b125435016207627
  • FukuenSIwakiMYasuiASulfonylurea agents exhibit peroxisome proliferator-activated receptor gamma agonistic activityJ Biol Chem200528023653915764598
  • FullertSSchneiderFHaakEEffects of pioglitazone in nondiabetic patients with arterial hypertension: a double-blind, placebo-controlled studyJ Clin Endocrinol Metab2002875503612466345
  • GeisenKVeghAKrauseECardiovascular effects of conventional sulfonylureas and glimepirideHorm Metab Res1996284965078911987
  • GerberPLubbenGHeuslerSEffects of pioglitazone on metabolic control and blood pressure: a randomised study in patients with type 2 diabetes mellitusCurr Med Res Opin200319532914594526
  • GersteinHCYusufSBoschJEffect of rosiglitazone on the frequency of diabetes in patients with impaired glucose tolerance or impaired fasting glucose: a randomised controlled trialLancet2006368109610516997664
  • GhoshSStandenNBGalinianesMFailure to precondition pathological human myocardiumJ Am Coll Cardiol200137711811693741
  • GilliesPSDunnCJPioglitazoneDrugs20006033343 discussion 344–510983737
  • GoldbergRBKendallDMDeegMAA comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemiaDiabetes Care20052815475415983299
  • GossetPCharbonnierASDelerivePPeroxisome proliferator-activated receptor gamma activators affect the maturation of human monocyte-derived dendritic cellsEur J Immunol20013128576511592060
  • HanefeldMBrunettiPSchernthanerGHOne-year glycemic control with a sulfonylurea plus pioglitazone versus a sulfonylurea plus metformin in patients with type 2 diabetesDiabetes Care200427141714693980
  • HellmanBSehlinJTaljedalIBGlibenclamide is exceptional among hypoglycaemic sulphonylureas in accumulating progressively in beta-cell-rich pancreatic isletsActa Endocrinol Copenh1984105385906230850
  • HenryRRThiazolidinedionesEndocrinol Metab Clin North Am199726553739314015
  • HerzMJohnsDReviriegoJA randomized, double-blind, placebo-controlled, clinical trial of the effects of pioglitazone on glycemic control and dyslipidemia in oral antihyperglycemic medication-naive patients with type 2 diabetes mellitusClin Ther20032510749512809958
  • InukaiKWatanabeMNakashimaYGlimepiride enhances intrinsic peroxisome proliferator-activated receptor-gamma activity in 3T3-L1 adipocytesBiochem Biophys Res Commun20053284849015694373
  • InzucchiSEOral antihyperglycemic therapy for type 2 diabetes: scientific reviewJAMA20022873607211790216
  • IRISThe IRIS III study: pioglitazone improves metabolic control and blood pressure in patients with type 2 diabetes without increasing body weightDiabetes, Obesity and Metabolism200791323
  • JainROseiKKupferSLong-term safety of pioglitazone versus glyburide in patients with recently diagnosed type 2 diabetes mellitusPharmacotherapy20062613889516999648
  • JernbergTJamesSLindahlBNatriuretic peptides in unstable coronary artery diseaseEur Heart J20042514869315342167
  • KahnSEHaffnerSMHeiseMAGlycemic durability of rosiglitazone, metformin, or glyburide monotherapyN Engl J Med200635524274317145742
  • KanataniYUsuiIIshizukaKEffects of pioglitazone on suppressor of cytokine signaling 3 expression: potential mechanisms for its effects on insulin sensitivity and adiponectin expressionDiabetes20075679580317327450
  • KaramJHMatinSBForshamPHAntidiabetic drugs after the University Group Diabetes Program UGDPAnnu Rev Pharmacol197515351661096790
  • KarthikeyanVJLipGYN-terminal pro-brain natriuretic peptide and coronary artery diseaseEur J Clin Invest2007371717181561
  • KawamoriRMatsuhisaMKinoshitaJPioglitazone enhances splanchnic glucose uptake as well as peripheral glucose uptake in non-insulin-dependent diabetes mellitus. AD-4833 Clamp-OGL Study GroupDiabetes Res Clin Pract19984135439768370
  • KhanMXuYEdwardsGEffects of pioglitazone on the components of diabetic dyslipidaemia: results of double-blind, multicentre, randomised studiesInt J Clin Pract2004589071215587767
  • KipnesMSKrosnickARendellMSPioglitazone hydrochloride in combination with sulfonylurea therapy improves glycemic control in patients with type 2 diabetes mellitus: a randomized, placebo-controlled studyAm J Med200111110711448655
  • KlepzigHKoberGMatterCSulfonylureas and ischaemic preconditioning; a double-blind, placebo-controlled evaluation of glimepiride and glibenclamideEur Heart J1999204394610213347
  • KramerWMullerGGirbigFDifferential interaction of glimepiride and glibenclamide with the beta-cell sulfonylurea receptor. II. Photoaffinity labeling of a 65 kDa protein by [3H]glimepirideBiochim Biophys Acta19941191278908172913
  • LangenfeldMRForstTHohbergCPioglitazone decreases carotid intima-media thickness independently of glycemic control in patients with type 2 diabetes mellitus: results from a controlled randomized studyCirculation200511125253115883215
  • LeeTMChouTFImpairment of myocardial protection in type 2 diabetic patientsJ Clin Endocrinol Metab200388531712574175
  • LehmannJMMooreLBSmith-OliverTAAn antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma PPAR gammaJ Biol Chem19952701295367768881
  • LehtihetMWelshNBerggrenPOGlibenclamide inhibits islet carnitine palmitoyltransferase 1 activity, leading to PKC-dependent insulin exocytosisAm J Physiol Endocrinol Metab2003285E4384612684219
  • LembergerTDesvergneBWahliWPeroxisome proliferator-activated receptors: a nuclear receptor signaling pathway in lipid physiologyAnnu Rev Cell Dev Biol199612335638970730
  • Luis BautistaJBugosCDirnbergerGEfficacy and safety profile of glimepiride in Mexican American patients with type 2 diabetes mellitus: a randomized, placebo-controlled studyClin Ther20032519420912637120
  • MartensFMRabelinkTJop’t RoodtJTNF-alpha induces endothelial dysfunction in diabetic adults, an effect reversible by the PPAR-gamma agonist pioglitazoneEur Heart J2006271605916762982
  • MartensFMVisserenFLde KoningEJShort-term pioglitazone treatment improves vascular function irrespective of metabolic changes in patients with type 2 diabetesJ Cardiovasc Pharmacol200546773816306801
  • MarxNMachFSautyAPeroxisome proliferator-activated receptor-gamma activators inhibit IFN-gamma-induced expression of the T cell-active CXC chemokines IP-10, Mig, and I-TAC in human endothelial cellsJ Immunol20001646503810843708
  • MarxNWohrleJNusserTPioglitazone reduces neointima volume after coronary stent implantation: a randomized, placebo-controlled, double-blind trial in nondiabetic patientsCirculation20051122792816246947
  • MattooVEcklandDWidelMMetabolic effects of pioglitazone in combination with insulin in patients with type 2 diabetes mellitus whose disease is not adequately controlled with insulin therapy: results of a six-month, randomized, double-blind, prospective, multicenter, parallel-group studyClin Ther2005275546715978304
  • MazzoneTMeyerPMFeinsteinSEffect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes: a randomized trialJAMA200629625728117101640
  • MiyazakiYGlassLTriplittCEffect of rosiglitazone on glucose and non-esterified fatty acid metabolism in Type II diabetic patientsDiabetologia2001a442210911793023
  • MiyazakiYMahankaliAMatsudaMImproved glycemic control and enhanced insulin sensitivity in type 2 diabetic subjects treated with pioglitazoneDiabetes Care2001b24710911315836
  • MiyazakiYMahankaliAMatsudaMEffect of pioglitazone on abdominal fat distribution and insulin sensitivity in type 2 diabetic patientsJ Clin Endocrinol Metab2002a8727849112050251
  • MiyazakiYMahankaliAWajcbergEEffect of pioglitazone on circulating adipocytokine levels and insulin sensitivity in type 2 diabetic patientsJ Clin Endocrinol Metab2004894312915356026
  • MiyazakiYMatsudaMDeFronzoRADose-response effect of pioglitazone on insulin sensitivity and insulin secretion in type 2 diabetesDiabetes Care2002b255172311874940
  • MocanuMMMaddockHLBaxterGFGlimepiride, a novel sulfonylurea, does not abolish myocardial protection afforded by either ischemic preconditioning or diazoxideCirculation20011033111611425777
  • NakamuraTMatsudaTKawagoeYEffect of pioglitazone on carotid intima-media thickness and arterial stiffness in type 2 diabetic nephropathy patientsMetabolism2004531382615375799
  • Neuschwander-TetriBABruntEMWehmeierKRImproved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-gamma ligand rosiglitazoneHepatology20033810081714512888
  • NicholsCGLedererWJAdenosine triphosphate-sensitive potassium channels in the cardiovascular systemAm J Physiol1991261H1675861750525
  • NishioKSakuraiMKusuyamaTA randomized comparison of pioglitazone to inhibit restenosis after coronary stenting in patients with type 2 diabetesDiabetes Care200629101616373904
  • NolanJJLudvikBBeerdsenPImprovement in glucose tolerance and insulin resistance in obese subjects treated with troglitazoneN Engl J Med19943311188937935656
  • PanzramGMortality and survival in type 2 non-insulin-dependent. diabetes mellitusDiabetologia198730123313556287
  • PavoIJermendyGVarkonyiTTEffect of pioglitazone compared with metformin on glycemic control and indicators of insulin sensitivity in recently diagnosed patients with type 2 diabetesJ Clin Endocrinol Metab20038816374512679450
  • PerezAKhanMJohnsonTPioglitazone plus a sulphonylurea or metformin is associated with increased lipoprotein particle size in patients with type 2 diabetesDiab Vasc Dis Res20041445016305056
  • PfutznerAHohbergCLubbenGPioneer study: PPARgamma activation results in overall improvement of clinical and metabolic markers associated with insulin resistance independent of long-term glucose controlHorm Metab Res2005a37510516138265
  • PfutznerAMarxNLubbenGImprovement of cardiovascular risk markers by pioglitazone is independent from glycemic control: results from the pioneer studyJ Am Coll Cardiol2005b4519253115963388
  • PhatakHMYinDDFactors associated with the effect-size of thiazolidinedione TZD. therapy on HbA1c: a meta-analysis of published randomized clinical trialsCurr Med Res Opin20062222677817076987
  • ProksPReimannFGreenNSulfonylurea stimulation of insulin secretionDiabetes200251Suppl 3S3687612475777
  • ProutTEA prospective view of the treatment of adult-onset diabetes with special reference to the University Group Diabetes Program and oral hypoglycemic agentsMed Clin North Am1971551065754946757
  • QuastUStephanDBiegerSThe impact of ATP-sensitive K+ channel subtype selectivity of insulin secretagogues for the coronary vasculature and the myocardiumDiabetes200453Suppl 3S1566415561904
  • QuayleJMNelsonMTStandenNBATP-sensitive and inwardly rectifying potassium channels in smooth musclePhysiol Rev19977711652329354814
  • RasouliNRaueUMilesLMPioglitazone improves insulin sensitivity through reduction in muscle lipid and redistribution of lipid into adipose tissueAm J Physiol Endocrinol Metab2005288E930415632102
  • RichterBBandeira-EchtlerEBergerhoffKPioglitazone for type 2 diabetes mellitusCochrane Database Syst Rev2006CD00606017054272
  • RivelineJPDanchinNLedruFSulfonylureas and cardiovascular effects: from experimental data to clinical use. Available data in humans and clinical applicationsDiabetes Metab2003292072212909809
  • RosenblattSMiskinBGlazerNBThe impact of pioglitazone on glycemic control and atherogenic dyslipidemia in patients with type 2 diabetes mellitusCoron Artery Dis2001124132311491207
  • RosenstockJEinhornDHershonKEfficacy and safety of pioglitazone in type 2 diabetes: a randomized, placebo-controlled study in patients receiving stable insulin therapyInt J Clin Pract200256251712074206
  • RosenstockJSamolsEMuchmoreDBGlimepiride, a new once-daily sulfonylurea. A double-blind placebo-controlled study of NIDDM patients. Glimepiride Study GroupDiabetes Care199619119498908379
  • RubinsHBRobinsSJCollinsDGemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study GroupN Engl J Med1999341410810438259
  • SaltielAROlefskyJMThiazolidinediones in the treatment of insulin resistance and type II diabetesDiabetes199645166198922349
  • SanyalAJMofradPSContosMJA pilot study of vitamin E versus vitamin E and pioglitazone for the treatment of nonalcoholic steatohepatitisClin Gastroenterol Hepatol2004211071515625656
  • SarafidisPABakrisGLProtection of the kidney by thiazolidinediones: an assessment from bench to bedsideKidney Int20067012233316883325
  • SatoTNishidaHMiyazakiMNakayaHEffects of sulfonylureas on mitochondrial ATP-sensitive K+ channels in cardiac myocytes: implications for sulfonylurea controversyDiabetes Metab Res Rev200622341716444778
  • ScherbaumWAGokeBMetabolic efficacy and safety of once-daily pioglitazone monotherapy in patients with type 2 diabetes: a double-blind, placebo-controlled studyHorm Metab Res2002345899512439788
  • SchernthanerGGrimaldiADi MarioUGUIDE study: double-blind comparison of once-daily gliclazide MR and glimepiride in type 2 diabetic patientsEur J Clin Invest2004a345354215305887
  • SchernthanerGMatthewsDRCharbonnelBEfficacy and safety of pioglitazone versus metformin in patients with type 2 diabetes mellitus: a double-blind, randomized trialJ Clin Endocrinol Metab2004b8960687615579760
  • SchwartzAVSellmeyerDEVittinghoffEThiazolidinedione use and bone loss in older diabetic adultsJ Clin Endocrinol Metab20069133495416608888
  • ShadidSJensenMDEffects of pioglitazone versus diet and exercise on metabolic health and fat distribution in upper body obesityDiabetes Care20032631485214578253
  • SmithSRDe JongeLVolaufovaJEffect of pioglitazone on body composition and energy expenditure: a randomized controlled trialMetabolism200554243215562376
  • StamlerJVaccaroONeatonJDDiabetes, other risk factors, and 12-yr cardiovascular mortality for men screened in the Multiple Risk Factor Intervention TrialDiabetes Care199316434448432214
  • StrattonIMAdlerAINeilHAAssociation of glycaemia with macrovascular and microvascular complications of type 2 diabetes UKPDS 35.: prospective observational studyBMJ20003214051210938048
  • TakagiTYamamuroATamitaKPioglitazone reduces neointimal tissue proliferation after coronary stent implantation in patients with type 2 diabetes mellitus: an intravascular ultrasound scanning studyAm Heart J2003146E512891212
  • TanMJohnsDGonzalez GalvezGEffects of pioglitazone and glimepiride on glycemic control and insulin sensitivity in Mexican patients with type 2 diabetes mellitus: A multicenter, randomized, double-blind, parallel-group trialClin Ther2004a266809315220012
  • TanMHJohnsDStrandJSustained effects of pioglitazone vs. glibenclamide on insulin sensitivity, glycaemic control, and lipid profiles in patients with Type 2 diabetesDiabet Med2004b218596615270789
  • TiikkainenMHakkinenAMKorsheninnikovaEEffects of rosiglitazone and metformin on liver fat content, hepatic insulin resistance, insulin clearance, and gene expression in adipose tissue in patients with type 2 diabetesDiabetes20045321697615277403
  • TranMTNavarMDDavidsonMBComparison of the glycemic effects of rosiglitazone and pioglitazone in triple oral therapy in type 2 diabetesDiabetes Care2006291395616732030
  • UKPDSEffect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes UKPDS 34. UK Prospective Diabetes Study UKPDS. GroupLancet1998a352854659742977
  • UKPDSIntensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes UKPDS 33. UK Prospective Diabetes Study UKPDS. GroupLancet1998b352837539742976
  • WallaceTMLevyJCMatthewsDRAn increase in insulin sensitivity and basal beta-cell function in diabetic subjects treated with pioglitazone in a placebo-controlled randomized studyDiabet Med2004215687615154941
  • WilcoxRBousserMGBetteridgeDJEffects of pioglitazone in patients with type 2 diabetes with or without previous stroke: results from PROactive PROspective pioglitAzone Clinical Trial In macroVascular Events 04Stroke2007388657317290029
  • WillsonTMLambertMHKliewerSAPeroxisome proliferator-activated receptor gamma and metabolic diseaseAnnu Rev Biochem2001703416711395411
  • WinklerKKonradTFullertSPioglitazone reduces atherogenic dense LDL particles in nondiabetic patients with arterial hypertension: a double-blind, placebo-controlled studyDiabetes Care20032625889412941723
  • YamadaKJiJJYuanHProtective role of ATP-sensitive potassium channels in hypoxia-induced generalized seizureScience20012921543611375491
  • Yki-JarvinenHThe PROactive study: some answers, many questionsLancet20053661241216214581
  • ZhangFSjoholmKZhangQPioglitazone acutely influences glucose-sensitive insulin secretion in normal and diabetic human isletsBiochem Biophys Res Commun2006351750517084385
  • ZingmanLVHodgsonDMBastPHKir6.2 is required for adaptation to stressProc Natl Acad Sci U S A200299132788312271142

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.