Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Cardiol. Jul 26, 2014; 6(7): 517-530
Published online Jul 26, 2014. doi: 10.4330/wjc.v6.i7.517
Antihypertensive drugs and glucose metabolism
Christos V Rizos, Moses S Elisaf, Department of Internal Medicine, School of Medicine, University of Ioannina, 45110 Ioannina, Greece
Author contributions: Rizos CV and Elisaf MS contributed equally to this paper.
Correspondence to: Moses S Elisaf, MD, FASA, FRSH, Professor, Department of Internal Medicine, School of Medicine, University of Ioannina, Ipiros, 45110 Ioannina, Greece. egepi@cc.uoi.gr
Telephone: +30-26510-07509 Fax: +30-26510-07016
Received: December 20, 2013
Revised: March 23, 2014
Accepted: May 14, 2014
Published online: July 26, 2014

Abstract

Hypertension plays a major role in the development and progression of micro- and macrovascular disease. Moreover, increased blood pressure often coexists with additional cardiovascular risk factors such as insulin resistance. As a result the need for a comprehensive management of hypertensive patients is critical. However, the various antihypertensive drug categories have different effects on glucose metabolism. Indeed, angiotensin receptor blockers as well as angiotensin converting enzyme inhibitors have been associated with beneficial effects on glucose homeostasis. Calcium channel blockers (CCBs) have an overall neutral effect on glucose metabolism. However, some members of the CCBs class such as azelnidipine and manidipine have been shown to have advantageous effects on glucose homeostasis. On the other hand, diuretics and β-blockers have an overall disadvantageous effect on glucose metabolism. Of note, carvedilol as well as nebivolol seem to differentiate themselves from the rest of the β-blockers class, being more attractive options regarding their effect on glucose homeostasis. The adverse effects of some blood pressure lowering drugs on glucose metabolism may, to an extent, compromise their cardiovascular protective role. As a result the effects on glucose homeostasis of the various blood pressure lowering drugs should be taken into account when selecting an antihypertensive treatment, especially in patients which are at high risk for developing diabetes.

Key Words: Hypertension, Glucose metabolism, Antihypertensive drugs

Core tip: Hypertension is a major contributor to the development and progression of cardiovascular disease. Increased blood pressure often coexists with insulin resistance. The various antihypertensive drugs have different effect on glucose metabolism. Indeed, angiotensin receptor blockers as well as angiotensin converting enzyme inhibitors have been associated with beneficial effects on glucose homeostasis. Calcium channel blockers are considered to have neutral metabolic effects. On the other hand, diuretics and β-blockers have an overall disadvantageous effect on glucose metabolism. As a result the metabolic effects of the various blood pressure lowering drugs should be taken into account when selecting an antihypertensive treatment.



INTRODUCTION

Hypertension is a growing epidemic affecting an important percentage of the population[1]. Hypertensive patients have increased risk for the development and progression of both microvascular and macrovascular complications. As a result the need for a comprehensive management of high blood pressure is essential.

Hypertension is strongly associated with risk factors that impair glucose homeostasis and is often presented as a component of the metabolic syndrome. Indeed, hypertension is related with obesity, insulin resistance as well as diabetes mellitus[2,3]. As a result, hypertensive patients have a 2.5-fold higher risk of type 2 diabetes mellitus (T2DM) onset compared with normotensive subjects[4]. The various classes of antihypertensive drugs have different effects on blood glucose metabolism. Indeed, antihypertensive agents, such as β-blockers and thiazide diuretics have been associated with negative effects on blood glucose in contrast to other classes, such as angiotensin receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACE-I). As a result, the treatment of hypertensive subjects should be carefully selected as to not further deteriorate an already at risk glucose homeostasis.

RESEARCH

We searched PubMed up to December 2013 using combinations of the following keywords: hypertension, glucose metabolism, glucose homeostasis, antihypertensive drugs, angiotensin converting enzyme inhibitors, ARBs, calcium channel blockers (CCB), β blockers, renin inhibitors, alpha blockers, diuretics. Major randomized controlled trials, original papers, review articles and case reports were included. The references of these articles were scrutinized for relevant articles. For articles not written in English, only the abstracts were considered. A minor limitation of this review is that our literature search was exclusively based on the PubMed database.

RENIN ANGIOTENSIN ALDOSTERONE SYSTEM

The renin angiotensin aldosterone system (RAAS) is strongly associated with glucose homeostasis. A number of studies have identified antihypertensive drugs that act by intervening in the RAAS as overall having beneficial effects on glucose metabolism.

Angiotensin converting enzyme inhibitors

The majority of clinical trials evaluating the effects of ACE-I on glucose metabolism have showed a positive outcome. Large clinical trials have revealed that ACE-I are associated with a lower incidence of new-onset T2DM in hypertensive subjects. The heart outcomes prevention evaluation (HOPE) study demonstrated the favorable influence of ramipril on cardiovascular (CV) disease (CVD) incidence in high risk patients[5]. Patients recruited were ≥ 55 years old, had a history of coronary artery disease, stroke, peripheral vascular disease, or diabetes plus at least one other CV risk factor [hypertension, elevated total cholesterol levels, low high-density lipoprotein cholesterol (HDL-C) levels, cigarette smoking, or documented microalbuminuria]. For a mean period of 5 years the HOPE trial randomized the above high-risk patients (n = 9279) to ramipril (10 mg/d) or placebo. Ramipril reduced new onset diabetes by 34% (P < 0.001 vs placebo)[5]. However, there are some limitations of the HOPE results regarding new onset diabetes. Indeed, diabetes development in HOPE was not a pre-specified endpoint of the study. Moreover, the diagnosis of diabetes was patient reported.

Similarly, the Captopril Prevention Project (CAPPP) study was a prospective, randomized trial which compared the effect of captopril vs antihypertensive treatment with diuretics, β-blockers, or both in hypertensive patients (n = 10985)[6]. Treatment with captopril was associated with fewer patients developing diabetes compared with the control group (OR = 0.79; 95%CI: 0.67-0.94; P = 0.007). However, because of the design of the study, the query arises as to whether the differences in development of T2DM in the CAPPP trial were due to a protective effect of ACE-I or a deleterious effect of β-blockers and diuretics. Another limitation of the study was that blood pressure as well as diabetes mellitus at baseline was more common in the captopril group than in the group that received conventional treatment. In addition, in the captopril group a diuretic or a CCB was added to treatment in order to achieve the blood pressure goal.

The Antihypertensive and Lipid-Lowering Treatment to prevent heart attack trial (ALLHAT) was a randomized, double-blind, trial which evaluated whether treatment with a CCB or an ACE-I lowers the incidence of coronary heart disease (CHD) or other CVD events vs treatment with a diuretic[7]. Patients (n = 33357) with hypertension and at least one other cardiac heart disease risk factor were randomized to chlorthalidone, amlodipine, or lisinopril for a mean follow-up of 4.9 years. Lisinopril treatment reduced the relative risk of developing T2DM by 30% (95%CI: 23%-37%; P < 0.001) compared with patients treated with chlorthalidone and by 17% (95%CI: 7%-26%; P < 0.01) compared with patients treated with the amlodipine[7].

The studies of left ventricular dysfunction (SOLVD) was a double-blind trial which randomized patients with asymptomatic left ventricular (LV) dysfunction to receive enalapril or placebo for a mean follow-up of 37.4 mo[8]. Enalapril significantly reduced the incidence of heart failure and the rate of related hospitalizations compared with placebo[8]. A retrospective study evaluated the effect of enalapril on the incidence of diabetes in patients from the SOLVD trial[9]. Enalapril significantly reduced the incidence of diabetes compared with placebo (HR = 0.22; 95%CI: 0.10-0.46; P < 0.0001)[9].

On the other hand, some studies have shown that ACE-I have a neutral effect on glucose metabolism. A study in patients with T2DM and hypertension (n = 24) resulted in no change in insulin sensitivity after trandolapril treatment[10]. Similarly, enalapril treatment did not affect insulin sensitivity in patients with essential hypertension (n = 20)[11]. Moreover, lisinopril did not affect insulin sensitivity in healthy volunteers (n = 22)[12]. The Diabetes Reduction Assessment with Ramipril and Rosiglitazone Medication (DREAM) study evaluated the effects of ramipril or placebo in patients (n = 5269) without CVD but with impaired fasting glucose levels or impaired glucose tolerance. Patients received ramipril (up to 15 mg per day) or placebo (and rosiglitazone or placebo) for a median of 3 years[13]. Although ramipril treatment did not reduce the incidence of diabetes, it increased regression to normoglycemia in the study population (P = 0.001). The Diabetes Reduction Assessment with Ramipril and Rosiglitazone Medication Ongoing Follow-up (DREAM On) study followed patients from the DREAM trial for a median 1.6 years after the end of the trial[14]. Ramipril did not influence diabetes occurrence. Similarly, regression to normoglycemia was not altered by ramipril.

A meta-analysis of randomized control trials associated ACE-I treatment with a reduction of new-onset T2DM (RR = 0.73; 95%CI: 0.63-0.84)[15]. Similar were the results of another meta-analysis of randomized clinical trials where ACE-I had a smaller incidence of new-onset T2DM (OR = 0.77; 95%CI: 0.72-0.82; P < 0.0006) compared with control groups[16].

ARBs

Treatment with ARBs has also been associated with an overall beneficial effect on glucose homeostasis. Indeed, large clinical trials have associated ARB treatment with lower incidence of new-onset T2DM. The losartan intervention for endpoint reduction (LIFE) in hypertension study was a double-blinded, randomized, parallel-group trial in patients (n = 9193) aged 55-80 years with essential hypertension (sitting blood pressure 160-200/95-115 mmHg) and LV hypertrophy[17]. Patients were randomized to losartan or atenolol based antihypertensive treatment for a mean follow-up of 4.8 years[17]. Losartan treatment was associated with a reduction of new-onset T2DM compared with the control group (HR = 0.75; 95%CI: 0.63-0.88; P = 0.001).

The Antihypertensive treatment and Lipid Profile In a North of Sweden Efficacy Evaluation (ALPINE) study compared the effect of hydrochlorothiazide, alone or in combination with atenolol, against candesartan, alone or in combination with felodipine, in newly diagnosed patients with primary hypertension (n = 342)[18]. After 12 mo, fasting plasma glucose and fasting serum insulin increased in the diuretic group, while a decrease was observed in the candesartan group (P < 0.001 for the comparison of the 2 groups). The incidence of new-onset T2DM was higher in the hydrochlorothiazide (4.1%) group compared with the candesartan group (0.5%; P = 0.03)[18].

The Valsartan Antihypertensive Long-term Use Evaluation (VALUE) was a prospective, double-blind, randomized trial that recruited hypertensive patients with additional CV risk factors[19]. Study subjects were randomized to either valsartan or amlodipine based regimen. Drug uptitration or the addition of further antihypertensive drugs, excluding ARBs, was allowed to achieve BP control. The valsartan based group had a smaller incidence of new-onset T2DM compared with the amlodipine group (HR = 0.77; 95%CI: 0.69-0.86; P < 0.0001)[19].

The Candesartan in Heart failure Assessment of Reduction in Mortality and morbidity (CHARM) study was a double-blind randomized control trial which evaluated candesartan vs placebo in patients with heart failure (n = 7599) for a median follow-up of 37.7 mo[20]. Among patients without a history of diabetes, new-onset T2DM was significantly lower in the candesartan group compared with the placebo group (HR = 0.78; 95%CI: 0.64-0.96; P = 0.020)[20]. The CHARM program consisted of 3 component trials, each comparing candesartan with placebo in a distinct population of patients with symptomatic heart failure: (1) the CHARM-Alternative which included patients with LV ejection fraction (LVEF) ≤ 40% and intolerant of ACE-I; (2) the CHARM-Added which included patients with LVEF ≤ 40% who were treated with an ACE-I; and (3) the CHARM-Preserved which included patients with LVEF > 40%. The candesartan group had a smaller incidence of T2DM compared with placebo only in the CHARM-Preserved trial (OR = 0.60; 95%CI: 0.41-0.86; P = 0.005).

The nateglinide and valsartan in impaired glucose tolerance outcomes research (NAVIGATOR) was a double-blind, randomized clinical trial in subjects with impaired glucose tolerance with known CVD or with CV risk factors[21]. Patients (n = 9518) were randomized to receive valsartan (up to 160 mg daily) or placebo for a median of 5.0 years. The valsartan group had a smaller incidence of T2DM compared with placebo (HR = 0.86; 95%CI: 0.80-0.92; P < 0.001)[21]. Despite the reduction of T2DM incidence, valsartan treatment did not reduce the rate of CV events.

On the other hand, the Study on Cognition and Prognosis in the Elderly (SCOPE) evaluated the effects of candesartan vs placebo in elderly patients aged 70-89 years (n = 4964) with hypertension for a mean follow-up of 3.7 years[22]. Open-label active antihypertensive therapy was added as needed. There was not a significant difference regarding new-onset T2DM between the 2 groups[22]. Similarly the CHARM-Added as well as the CHARM-Alternative studies did not show a difference regarding new-onset T2DM with candesartan treatment[20].

A number of meta-analyses indicate the protective role of ARB treatment regarding T2DM development. Geng et al[23] in a meta-analysis of 11 randomized control trials with 79773 patients (59862 non-diabetic patients at baseline) showed a beneficial effect of ARBs on T2DM development. Incidence of new-onset diabetes was significantly reduced in the ARBs group compared with controls (OR = 0.79; 95%CI: 0.74-0.84). This reduction of T2DM incidence was apparent in the comparison of ARBs to placebo (OR = 0.83; 95%CI: 0.78-0.89), β-blockers (OR = 0.73; 95%CI: 0.62-0.87), CCBs (OR = 0.76; 95%CI: 0.68-0.85) and non-ARBs (OR = 0.57; 95%CI: 0.36-0.91)[23]. ARBs were associated with significant reduction in the risk of new-onset diabetes in patients with hypertension (OR = 0.74; 95%CI: 0.68-0.81), heart failure (OR = 0.70; 95%CI: 0.50-0.96), impaired glucose tolerance (OR = 0.85; 95%CI: 0.78-0.92) or cardiocerebrovascular diseases (OR = 0.84; 95%CI: 0.72-0.97). A meta-analysis by Abuissa et al[15] of randomized controlled trials associated ARBs treatment with a reduction of new-onset T2DM [RR = 0.77 (95%CI: 0.71-0.83)][15]. Another meta-analysis of randomized clinical trials showed that ARBs had a smaller risk of new-onset T2DM (OR = 0.79; 95%CI: 0.73-0.85; P < 0.00001) compared with control groups[16]. Similarly, Cheung et al in a meta-analysis of studies with ARBs showed that sartans were associated with a decrease of new-onset diabetes[24].

Telmisartan: Among members of the ARB family, some have the ability to partially activate PPARγ. Indeed, when various ARBs were evaluated regarding their PPARγ activating capacity, telmisartan was identified as the most prominent one[25,26]. Irbesartan was also associated with a milder activation of PPARγ. However only telmisartan retained its PPARγ-activating ability in lower concentrations usually attained during oral drug treatment[25]. This capacity of telmisartan can be attributed, at least partially, to its unique structure which differentiates it from other ARBs as well as to its structural resemblance with pioglitazone, a full PPARγ agonist[25]. Telmisartan in contrast to thiazolidinediones is only a partial PPARγ agonist. This leads to a diverse but overlapping gene expression compared with full activation of PPARγ and thus bestowing upon telmisartan unique pleiotropic effects[27].

A number of studies have identified telmisartan as having beneficial effects on glucose homeostasis both in non-diabetic subjects[28,29] as well as diabetic patients[30,31]. Furthermore, studies comparing telmisartan with other ARBs have shown that telmisartan had more favorable effects on glycemic profile[32,33]. Hypertension often co-exists with dyslipidemia as commonly seen in metabolic syndrome. Moreover, there have been studies associating statin treatment with deteriorating effects on glucose metabolism[34-37]. Indeed, in the Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin trial (JUPITER)[38] rosuvastatin was associated with an increase in physician-reported newly diagnosed diabetes (P = 0.01) and an increase in glycated hemoglobin (HbA1c) vs placebo (P = 0.001). We have shown that telmisartan not only retains its beneficial effects on glucose homeo=stasis when co-administered with a statin, but also seems to negate any adverse effect of statin therapy on glycemic indices[39]. Patients (n = 151) with mixed dyslipidemia, stage 1 hypertension and prediabetes were randomized to receive rosuvastatin (10 mg/d) plus telmisartan 80 mg/d or irbesartan 300 mg/d or olmesartan 20 mg/d[39]. After 6 mo, the homeostasis Model Assessment Insulin Resistance (HOMA-IR) index improved only in the telmisartan group (-29%) compared with either irbesartan (+16%; P < 0.01 vs RT) or olmesartan group (+14%; P < 0.05 vs RT) (P < 0.05 for all vs baseline).

A number of large clinical trials have evaluated the effect of telmisartan on the incidence of new-onset T2DM. The Ongoing Telmisartan Alone and in Combination with Ramipril Global Endpoint Trial (ONTARGET) evaluated the effects of telmisartan on hard clinical endpoints[40]. High risk patients (n = 25620) with coronary, peripheral or cerebrovascular disease or diabetes with end-organ damage were randomized to 3 groups and were followed for a median period of 56 mo. The first group received telmisartan (80 mg/d), the second group ramipril (10 mg/d) and the third group telmisartan plus ramipril (80/10 mg/d). The ONTARGET trial did not reveal any difference between ramipril (6.7%) and telmisartan (7.5%; HR = 1.12; 95%CI: 0.97-1.29) regarding new onset diabetes[40].

The Telmisartan Randomised AssessmeNt Study in ACE iNtolerant subjects with CVD (TRANSCEND) came as a complementary study to ONTARGET[41]. High risk patients (n = 5926) intolerant to ACE-I with coronary, peripheral or cerebrovascular disease or diabetes with end-organ damage were randomized to telmisartan (80 mg/d) or placebo on top of any current therapy. TRANSCEND had the same primary endpoint as ONTARGET. A clear trend in reducing new clinical diagnosis of diabetes with telmisartan was seen in the TRANSCEND trial. The telmisartan group had lower new diabetes incidence (11%) vs placebo (12.8%; P = 0.081).

The prevention regimen for effectively avoiding second strokes (PRoFESS) study evaluated the effects of telmisartan on stroke incidence after a mean period of 30 mo[42]. Patients (n = 20332) with a history of recent ischemic stroke were randomly assigned (2 × 2) to receive either both aspirin (25 mg/twice daily) and extended-release dipyridamole (200 mg/twice daily) or clopidogrel (75 mg/d); and telmisartan (80 mg/d) or placebo. Similarly, in the PRoFESS a trend was seen in reducing new onset diabetes with telmisartan (1.2%) vs placebo (1.5%; P = 0.1).

Although the TRANSCEND and PRoFESS both only showed trends for the reduction of new-onset T2DM, it should be noted that both of them had some limitations regarding their power to identify beneficial effects of telmisartan on diabetes onset. Indeed, more than one third of the TRANSCEND population had already a history of diabetes, thus decreasing the power of the remaining study population to detect any mild beneficial effect on T2DM development. Moreover, a great percentage (37%) of the PRoFESS population was already treated with ACE-Is, which have an established overall positive effect regarding new onset diabetes prevention[43]. Therefore, again any benefits of telmisartan would be harder to detect on-top of an ACE-I therapy. Moreover, the PRoFESS had a much smaller follow up period in contrast to studies with ARBs that showed benefits in new onset diabetes like the LIFE[17] and VALUE[19], Indeed, the PRoFESS population was monitored for 2.5 years vs 4.8 and 4.2 years for the LIFE and VALUE populations, respectively. This difference could explain why telmisartan showed only a trend for reduction of new onset diabetes.

Renin inhibitors

Aliskiren is the first approved renin inhibitor which acts by directly inhibiting the renin enzyme at the point of RAAS activation, blocking the conversion of angiotensinogen to angiotensin I and decreasing levels of angiotensin I and angiotensin II[44]. A limited number of studies have evaluated the capacity of aliskiren to affect glucose metabolism. In a recent study, hypertensive patients with abnormal LV diastolic dysfunction but with normal LV systolic function (n = 78) were randomized to aliskiren (up to 300 mg/d) treatment or control group which was treated with β-blockers or CCBs[45]. Fasting insulin and glucose remained unchanged in the aliskiren group, in contrast to the control group where an increase in both fasting insulin (P = 0.03) and glucose (P = 0.003) were observed. In another double-blind trial, patients with diabetes mellitus and hypertension (n = 837) were randomized to once-daily aliskiren (150 mg titrated to 300 mg after four weeks), ramipril (5 mg titrated to 10 mg) or the combination for eight weeks[46]. No changes in HbA1c and fasting plasma glucose were observed in any treatment group. Another study randomized hypertensive patients with metabolic syndrome to aliskiren (300 mg/d) or losartan (100 mg/d)[47]. At study end patients performed an euglycemic hyperinsulinemic clamp and insulin sensitivity was assessed by glucose infusion rate. Insulin resistance improved only in the aliskiren group compared with losartan group (P < 0.05 between groups).

Mechanisms

The RAAS plays a major role both in the pathogenesis of hypertension as well as glucose homeostasis. As a result, a number of mechanisms have been suggested that can play a role in the overall beneficial effect that drugs which effect RAAS have on glucose metabolism.

Bradykinin may play an important role towards a beneficial effect on glucose homeostasis. The ACE beyond the conversion of angiotensin I to angiotensin II can also decrease bradykinin levels[48]. Indeed, ACE promotes the degradation of bradykinin to inactive fragments via a kininase II - mediated mechanism[49]. As a result, ACE-I can increase bradykinin levels[50]. Bradykinin has been shown to promote insulin sensitivity at the skeletal muscle level[51,52].

The principal glucose transporter protein that mediates insulin-stimulated glucose transport into muscle and adipose tissues is the glucose transporter type 4 (GLUT4), thus playing a key role in the regulation of glucose homeostasis[53]. Angiotensin II decreases GLUT-4 translocation to the cell membrane[54,55]. As a result the RAAS inhibition could promote insulin sensitivity. Indeed, the inhibition of AT1 receptors prevented the decline of GLUT-4 in a diabetic rat heart model[56]. Moreover, both ACEIs and ARBs have been associated with increase of GLUT-4 protein expression in skeletal muscle and myocardium in insulin-resistant animal models[57].

Moreover, angiotensin II inhibits adipogenic differentiation of human adipocytes via the AT1 receptor[58]. Angiotensin II may inhibit preadipocytes recruitment, resulting in the storage of lipids in muscle and other tissues, thus increasing insulin resistance[59]. As a result, the blockade of RAAS would promote the recruitment of preadipocytes thereby increasing the number of small insulin-sensitive adipocytes leading to improved insulin sensitivity.

Furthermore, angiotensin II can promote the production of inflammatory cytokines[60]. Inflammatory cytokines promote oxidative stress thus also leading to increased insulin resistance. In addition, endothelial dysfunction is also associated with insulin resistance[61]. Angiotensin converting enzyme inhibitors have also been shown to improve vascular function and insulin-mediated vascular responses[61]. Furthermore, ACE-I may also have direct beneficial effects on pancreatic β cells[62].

In addition ACE inhibition can lead to vasodilation of blood vessels[63]. This vasodilation has as a result the increment of total perfused area and thus increases glucose uptake and insulin sensitivity[64,65]. The activation of the sympathetic nervous system has also been associated with insulin resistance[66]. Both ACE-I[67] as well as ARBs[68] have been shown to decrease levels of catecholamines such as norepinephrine and epinephrine, thus further contributing to amelioration of insulin resistance.

Potassium levels play a significant role in insulin secretion since hypokalemia substantially impairs the insulin secretory response to glucose. As a result the increase of potassium levels by inhibiting the RAAS may also contribute to the improvement of glucose levels. Moreover, magnesium has also been shown to affect glucose homeostasis. Indeed, magnesium deficiency is associated with both a reduced cellular insulin action[69] and impaired insulin secretion[70]. The inhibition of the RAAS system leads to increased magnesium levels. A pooled analysis of studies using ACEIs in patients (n = 96) with essential hypertension found that changes in insulin sensitivity index (ISI) were directly correlated to alterations in serum magnesium levels[71].

CCBS

CCBs are generally considered as having an overall neutral metabolic profile. Indeed, a recent meta-analysis of 10 randomized clinical trials evaluated the effect of CCB treatment on new onset T2DM[72]. The overall risk of diabetes among subjects taking CCBs was not significant (RR = 0.99; 95%CI: 0.85-1.15). Compared with other classes of antihypertensive drugs, CCBs were associated with a higher incidence of diabetes than ACEIs (pooled risk ratio 1.23; 95%CI: 1.01-1.51) or ARBs (1.27; 95%CI: 1.14-1.42) and a lower incidence compared with β-blockers (RR = 0.83; 95%CI: 0.73-0.94) or diuretics (RR = 0.82; 95%CI: 0.69-0.98).

Another recent meta-analysis of 5 clinical trials compared the efficacy of ARBs and CCBs regarding their effect on insulin resistance as assessed using the HOMA-IR index in non-diabetic patients[73]. Both ARBs and CCBs had a similar effect on blood pressure reduction. However, ARBs reduced the HOMA-IR index (weighted mean difference -0.65, 95%CI: -0.93--0.38) and fasting plasma insulin (weighted mean difference -2.01, 95%CI: -3.27--0.74) significantly more than CCBs. A recent reanalysis of data from the NAVIGATOR trial showed that CCBs were not associated with new onset diabetes (HR = 0.95; 95%CI: 0.79-1.13)[74].

Of note overdose of CCB has been associated with hyperglycemia primarily due to the blockade of pancreatic L-type calcium channels and insulin resistance on the cellular level[75].

However, not all members of the CCB class have the same effect on glucose homeostasis. Indeed, azelnidipine has been associated with beneficial effect on glucose homeostasis in a diabetic animal model[76]. Moreover, similar beneficial effects were seen in a small study in non-diabetic patients (n = 17) with essential hypertension who had controlled blood pressure levels using amlodipine (5 mg/d)[77]. Azelnidipine (16 mg/d) or amlodipine (5 mg/d) was administered in a crossover design for 12-wk. Despite similar blood pressure reduction, azelnidipine significantly decreased levels of glucose and insulin 120 min after the 75 g oral glucose tolerance test (OGTT) (P < 0.05 vs amlodipine). This effect may be associated with the anti-inflammatory effects of azalnidipine[78], since proinflammatory cytokines have been associated with impaired glucose tolerance[79]. Furthermore, azelnidipine inhibits the enhancement of sympathetic nervous activity[80]. Since the activation of the sympathetic nervous system has been associated with insulin resistance[66], azalnidipine treatment may contribute to the amelioration of insulin resistance.

Another interesting member of the CCB class is manidipine[81]. A beneficial effect on insulin resistance has been shown with manidipine treatment[82]. The beneficial effects of manidipine have been observed in both non-diabetic and T2DM patients[83,84]. Furthermore, we have recently shown that manidipine can ameliorate the possible statin-associated increase in insulin resistance[85]. In a prospective, randomized, open-label, blinded endpoint study a total of 40 patients with impaired fasting glucose, mixed dyslipidemia, and stage 1 hypertension were included. Patients were randomly allocated to rosuvastatin (10 mg/d) plus olmesartan (20 mg/d) or manidipine (20 mg/d). After 3 mo, a significant increase in HOMA-IR index by 14% (P = 0.02 vs baseline) was seen in the olmesartan plus rosuvastatin group. On the other hand, HOMA-IR index did not change in the manidipine plus rosuvastatin group (P = NS vs baseline; P = 0.04 vs olmesartan plus rosuvastatin group). This favorable effect of manidipine may be linked to the drug’s capacity to partially activate the PPARγ which plays a major role in glucose metabolism[82]. Indeed, the effect of manidipine to activate PPARγ is about two-thirds of that of pioglitazone, a full PPARγ activator[82]. This partial activation of PPARγ may contribute to the avoidance of side effects commonly associated with thiazolidinediones treatment. Moreover, an increase of adiponectin levels (which are inversely associated with the development of insulin resistance and metabolic syndrome) has been observed with manidipine[86]. Furthermore, manidipine induces a smaller activation of the sympathetic nervous system, which can also play a role in the beneficial effects on glucose homeostasis. Indeed, when compared with other CCBs, manidipine is associated with lower levels of plasma norepinephrine[87].

β-BLOCKERS

A number of studies have associated treatment with β-blockers as having a disadvantageous effect on glucose homeostasis[88-91]. Indeed, a prospective study of three cohorts, namely the Nurses’ Health Study (NHS) I and II and the Health Professionals Follow-up Study evaluated the association between the use of different classes of antihypertensive medications and the risk of T2DM incident[92]. Treatment with a β-blocker was associated with a greater risk for the development of diabetes. Similarly, in the Atherosclerosis Risk in Communities study β-blockers led to an increase of risk for new-onset T2DM (RR = 1.28; 95%CI: 1.04-1.57)[4]. A large meta-analysis of patients with hypertension (n = 94492) treated with beta blockers evaluated the risk for the development of T2DM[93]. Beta-blocker therapy resulted in a 22% increased risk for new-onset T2DM (RR = 1.22, 95%CI: 1.12-1.33) compared with non-diuretic antihypertensive agents. On the other hand, a recent reanalysis of data from the NAVIGATOR trial showed that β-blockers were not associated with new onset diabetes (HR = 1.10, 95%CI: 0.92-1.31)[74].

However, not all members of the β blocker class have similar effect on glucose homeostasis. Indeed, carvedilol as well as nebivolol have shown a differentiation from the rest of the class[94,95]. The Glycemic Effects in Diabetes Mellitus: Carvedilol-Metoprolol Comparison in Hypertensives (GEMINI) study was a randomized, double-blind, parallel-group trial that compared the effects of carvedilol and metoprolol tartrate on glycemic control[96]. Patients (n = 1235) with hypertension (> 130/80 mmHg) and T2DM that were already receiving RAS blockers were randomized to receive carvedilol (6.25-25 mg/twice daily) or metoprolol (50-200 mg/twice daily). Open-label hydrochlorothiazide and a dihydropyridine calcium antagonist were added, if needed, to achieve blood pressure target. While blood pressure control was similar between groups, a difference was seen regarding glucose effects. The HbA1c increased with metoprolol (by 0.15%; P < 0.001) but not carvedilol (by 0.02%; P = 0.65). Moreover, insulin sensitivity improved with carvedilol (9.1%; P = 0.004) but not metoprolol (2.0%; P = 0.48 vs baseline; P = 0.004 between groups). Similarly, a study in subjects with metabolic syndrome compared nebivolol (5 mg/d) with metoprolol (100 mg/d)[97]. After 12-wk treatment both nebivolol and metoprolol had similarly decreased blood pressure and heart rate. However, metoprolol decreased insulin sensitivity compared with nebivolol (P = 0.03).

Mechanisms

Several possible mechanisms that may be responsible for the disadvantageous effect of β-blockers have been described. Treatment with conventional β-blockers leads to an unopposed a1-activity which causes vasoconstriction and decreased blood flow to the muscles, which are an important organ in the regulation of glucose homeostasis[98,99]. As a result a decrease in insulin-stimulated glucose uptake would occur, leading to insulin resistance. Furthermore, β-blockers can also decrease the first phase of insulin secretion from pancreatic β cells[88,89]. In addition, treatment with β-blockers can also lead to weight gain[100]. Since increased body weight is strongly associated with insulin resistance[101], this effect of β-blockers can further deteriorate glucose homeostasis.

DIURETICS

An important class of antihypertensive drugs is diuretics. This class includes loop diuretics such as furosemide, thiazide diuretics such as hydrochlorothiazide, thiazide-like diuretics such as chlorthalidone and potassium-sparing diuretics, such as amiloride, eplerenone and spironolactone.

A number of studies have associated diuretic treatment of hypertension as having a negative effect on glucose homeostasis[18,102]. Indeed, a meta-analysis of 22 clinical trials with 143153 nondiabetic patients evaluated the effects of various antihypertensive drug classes on diabetes incidence[43]. Treatment with diuretic was associated with increased risk for new onset diabetes compared with other antihypertensive treatments as well as placebo[43]. A long-term cohort study with initially untreated hypertensive subjects (n = 795) evaluated new-onset diabetes incidence according to antihypertensive treatment[103]. Diuretic treatment was present in 53.5% of subjects that developed T2DM, compared with 30.4% of patients that did not develop diabetes (P = 0.002). Moreover, diuretic treatment was an independent predictor of new onset diabetes (P = 0.004). Furthermore, a recent reanalysis of data from the NAVIGATOR trial showed that diuretics were associated with an increased risk of new onset diabetes (HR = 1.23, 95%CI: 1.06-1.44)[74].

A post hoc subgroup analyses of the ALLHAT study among nondiabetic participants of the study who were randomized to receive chlorthalidone (n = 8419), amlodipine (n = 4958), or lisinopril (n = 5034) evaluated the effects of antihypertensive treatment on glucose levels as well as new-onset diabetes[104]. Chlorthalidone treatment was associated with a greater risk for developing diabetes compared with the other 2 treatment regimens (P < 0.001)[104]. The Systolic Hypertension in the Elderly Program (SHEP) was a placebo-controlled, double-blind, randomized, multicenter clinical trial that evaluated the efficacy of chlorthalidone in patients (n = 4736) with isolated systolic hypertension[105]. After 3 years of treatment, the incidence of new-onset diabetes was similar between the chlorthalidone (8.6%) and placebo group (7.5%; P = 0.25 between groups)[105]. However, when study participants were re-evaluated after a mean follow-up of 14.3 years, 13.0% of patients developed diabetes in the chlorthalidone group vs 8.7% in the placebo group (P < 0.0001)[106].

Of note, chlorthalidone seems to be differentiated from the rest of the thiazide diuretics class[107]. Indeed, chlorthalidone has a different chemical structure compared with the rest of thiazide diuretics[107] as well as the ability to inhibit carbonic anhydrase[108]. Carbonic anhydrase regulates a number of CV related risk factors[109,110] and its activity is also directly proportional to increasing blood glucose concentration[111]. As a result, chlorthalidone may have a more favorable metabolic profile compared with the other thiazide diuretics[107].

The effects of amiloride on blood glucose levels were evaluated in a study by Stears et al[112]. Patients with essential hypertension (n = 37) received, in random order, 4 wk of once-daily treatment with hydrochlorothiazide (25-50 mg), nebivolol (5-10 mg), combination (hydrochlorothiazide 25-50 mg and nebivolol 5-10 mg), amiloride (10-20 mg), and placebo. Each drug was force titrated at 2 wk and separated by a 4-wk placebo washout. Both amiloride and hydrochlorothiazide had similar changes in blood pressure reduction. However, an increase of glucose levels after a 2 h OGTT was observed with hydrochlorothiazide treatment, while no change was seen with amiloride (P < 0.0001).

The Eplerenone in Mild Patients Hospitalization and Survival Study in Heart Failure (EMPHASIS-HF) evaluated the effects of eplerenone on new-onset diabetes mellitus in patients (n = 1846) with mild heart failure[113]. After a follow-up of 21 mo, eplerenone had no effect on new-onset diabetes mellitus (HR = 0.94, 95%CI: 0.59-1.52). Another study compared the effects of eplerenone with spironolactone in patients (n = 107) with mild chronic heart failure[114]. Spironolactone increased levels of HbA1c (P < 0.0001), while no change was observed in the eplerenone group.

Mechanisms

Among the possible mechanisms through which thiazide diuretics may affect glucose homeostasis, hypokalemia may play an important role[115]. Indeed, hypokalemia can lead to decreased insulin secretion by β cells in response to glucose, as well as decrease in blood flow in muscles. A quantitative review evaluating studies that used thiazide diuretics, found an inverse relationship between glucose and potassium with thiazide use[116]. Similar results were observed in an analysis of data from the SHEP study[117]. In the first year of the study among 3790 nondiabetic participants each 0.5-mEq/L decrease in serum potassium was independently associated with a 45% higher adjusted diabetes risk (95%CI: 24%-70%; P < 0.001). However, a prespecified subgroup analysis of metabolic parameter data from patients participating in the Pharmacogenomic Evaluation of Antihypertensive Responses (PEAR) study did not confirm a relationship between hypokalemia and deterioration of serum glucose levels[118].

Moreover, a decrease in magnesium can be seen with diuretic treatment. This could also contribute to the disadvantageous effects of diuretics on glucose homeostasis, since hypomagnesaemia is an independent predictor of T2DM[119,120]. Furthermore, thiazide treatment is also associated with visceral fat redistribution, liver fat accumulation and low-grade inflammation, which in turn increase the risk of new-onset diabetes[121].

OTHER ANTIHYPERTENSIVE DRUGS

There is little evidence about the effects of other, less used, antihypertensive drugs on glucose homeostasis. A randomized, double-blind multicenter study compared moxonidine (0.2-0.6 mg/d) with metoprolol (50-150 mg/d) in hypertensive subjects (n = 127) with T2DM[122]. After 12 wk of treatment both groups had similar blood pressure reductions as well as similar HbA1c values. However, fasting plasma glucose decreased in the moxonidine group, while an increase was seen in the metoprolol group (P < 0.05). Furthermore, the HOMA-IR decreased with moxonidine in contrast to the increase observed with metoprolol. Another multicenter, prospective, randomized study compared moxonidine with metformin[123]. Patients older than 40 years old, with impaired glucose tolerance (or diabetes mellitus treated with diet alone) and a body mass index (BMI) of at least 27 kg/m2 were treated twice daily with moxonidine 0.2 mg or metformin 500 mg for 16 wk. Compared with metformin, moxonidine decreased the area under the curve for insulin (P = 0.049). On the other hand, only metformin significantly decreased fasting plasma glucose (P < 0.05 vs baseline and vs moxonidine) as well as HbA1c (P < 0.005 vs baseline). Both treatments similarly increased the Matsuda ISI from baseline to a comparable degree (P < 0.05 vs baseline for both groups). Another randomized open parallel study evaluated the chronic effects of moxonidine vs amlodipine in obese hypertensive patients (n = 40)[124]. Plasma levels of insulin 120 min after glucose load, decreased with moxonidine treatment (P < 0.05) while no change was seen with amlodipine. A multinational, open-label, observational study, the Moxonidine Efficacy on blood pressure Reduction revealed in a metabolic SYndrom population (MERSY) study evaluated the effects of moxonidine on serum metabolic parameters[125]. Patients with hypertension received moxonidine (0.2-0.4 mg/d) either as monotherapy or as adjunctive therapy for 6 mo. A beneficial trend in metabolic parameters such as fasting plasma glucose and body weight was observed with moxonidine.

A small study evaluated the effects of doxazosin in hypertensive non-insulin depended diabetic patients[126]. Doxazosin significantly improved insulin sensitivity during the euglycemic insulin clamp and enhanced OGTT. Similarly another small study showed a beneficial effect of doxazosin (2 mg or 4 mg daily for 3 mo) on insulin resistance indices in hypertensive patients (n = 21) with T2DM.

CONCLUSION

Hypertension is associated with increased morbidity and mortality. Furthermore, hypertensive patients have an increased prevalence of insulin resistance as often is the case with metabolic syndrome subjects. This disturbance in glucose homeostasis further increases the risk for the development of CVD as well as the development of diabetes. The various antihypertensive drug categories have different effects on glucose metabolism. Indeed, ACE-I and ARBs have the most favorable effect on insulin resistance and the development of T2DM. Moreover, CCBs have an overall neutral metabolic effect. However, both azelnidipine and manidipine have been associated with beneficial glucose effects. On the other hand, diuretics as well as β-blockers have been associated with detrimental effects on glucose metabolism.

An interesting query is whether the adverse effects of some antihypertensive drug categories on glucose metabolism and their potency to increase new-onset diabetes mellitus incidence is also associated with an increase in CVD events. It would be reasonable to assume that the drug-induced increases in glucose levels and T2DM incidence would have increased CVD risk similarly to traditional risk factors for new-onset diabetes. However, no such increase in CVD risk was seen in the ALLHAT study in those who developed diabetes in the chlorthalidone treatment arm[127]. Similarly were the results from the SHEP study[106]. Diabetes at baseline was associated with increased CV mortality rate (adjusted HR = 1.659, 95%CI: 1.413-1.949) and total mortality rate (adjusted HR = 1.510, 95%CI: 1.347-1.693). Furthermore, diabetes that developed during the trial among subjects on placebo was also associated with increased CV adverse outcome (adjusted HR = 1.562, 95%CI: 1.117-2.184) and total mortality rate (adjusted HR = 1.348, 95%CI: 1.051-1.727). However, diabetes that developed among subjects during diuretic therapy did not have statistically significant associations with CV mortality rate (adjusted HR = 1.043, 95%CI: 0.745-1.459) or total mortality rate (adjusted HR = 1.151, 95%CI: 0.925-1.433). In addition, diuretic treatment in diabetic patients was strongly associated with lower long-term CV mortality rate (adjusted HR = 0.688, 95%CI: 0.526-0.848) and total mortality rate (adjusted HR = 0.805, 95%CI: 0.680-0.952). Of note, even if new-onset T2DM after diuretic or β-blocker is not associated with increased CVD morbidity and mortality, the health care cost should be considered. Indeed, the management and treatment costs of a hypertensive patient with diabetes are far greater compared with a non-diabetic patient.

On the other hand, the Progetto Ipertensione Umbria Monitoraggio Ambulatoriale study, a long-term cohort study in initially untreated hypertensive subjects with a median follow up of 6 years, identified diuretic treatment as an independent predictor of new onset diabetes (P = 0.004)[103]. Of interest, CV event risk was similar between diabetic subjects at study baseline and subjects that developed new-onset T2DM during the study. An interesting study, evaluated hypertensive subjects (n = 754) and followed them long term for 25-28 years[128]. Patients were treated with thiazide diuretics and beta-adrenergic blocking drugs with the addition of hydralazin during the first decade. Calcium antagonists were substituted for hydralazin and, if needed, ACE-I were added when these drugs became available. After 25 years, treatment with β-blockers was associated with new-onset T2DM. New-onset diabetes was associated with an increased risk for stroke (HR = 1.67; 95%CI: 1.1-2.6; P < 0.05), myocardial infarction (OR = 1.66; 95%CI: 1.1-2.5; P < 0.05) and mortality (OR = 1.42; 95%CI: 1.1-1.9; P < 0.05). The mean observation time from onset of diabetes mellitus to a first stroke was 9.1 years and to a first myocardial infarction 9.3 years.

Despite the various effects of different antihypertensive drugs on glucose homeostasis, the overall expected benefits vs the potential risks should always be carefully weighted for each individual patient. As a result, when the benefits for a patient that should receive a treatment with an antihypertensive class with unfavorable glucose profile are greater than the risk of increased insulin resistance, then the glycemic effects of the antihypertensive drug should not disqualify the patient from treatment. Furthermore, there is often some diversity among the members of an antihypertensive class regarding their effect on glucose. As a result, the antihypertensive drug with the least adverse effect on glucose can be selected. Indeed, despite the overall adverse effect of the β-blockers families on glucose homeostasis, newer members of the class, such as carvedilol and nebivolol, have shown that they are clearly different from the rest regarding glucose effects.

Overall, when treating hypertensive patients the physician should carefully assess the individual patient’s medical history which often dictates a particular treatment. When there are no contraindications, an antihypertensive drug with a favorable or at least neutral effect on glucose homeostasis should be selected. This way, any beneficial effects of lowering blood pressure would not be shadowed in any way by a worsening of the metabolic profile. Patients with a strong indication for receiving a β-blocker or a diuretic should not be disqualified only because of the negative effect of these drug categories on glucose homeostasis. When a drug with negative effects on glucose homeostasis is selected, the physician should have in mind the possible deterioration of glucose metabolism and increased risk for new-onset diabetes and thus follow-up the patient accordingly.

Footnotes

P- Reviewer: Leone A S- Editor: Song XX L- Editor: A E- Editor: Wu HL

References
1.  Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet. 2005;365:217-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2858]  [Cited by in F6Publishing: 3634]  [Article Influence: 191.3]  [Reference Citation Analysis (1)]
2.  Lind L, Berne C, Lithell H. Prevalence of insulin resistance in essential hypertension. J Hypertens. 1995;13:1457-1462.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Lender D, Arauz-Pacheco C, Adams-Huet B, Raskin P. Essential hypertension is associated with decreased insulin clearance and insulin resistance. Hypertension. 1997;29:111-114.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Gress TW, Nieto FJ, Shahar E, Wofford MR, Brancati FL. Hypertension and antihypertensive therapy as risk factors for type 2 diabetes mellitus. Atherosclerosis Risk in Communities Study. N Engl J Med. 2000;342:905-912.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med. 2000;342:145-153.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Hansson L, Lindholm LH, Niskanen L, Lanke J, Hedner T, Niklason A, Luomanmäki K, Dahlöf B, de Faire U, Mörlin C. Effect of angiotensin-converting-enzyme inhibition compared with conventional therapy on cardiovascular morbidity and mortality in hypertension: the Captopril Prevention Project (CAPPP) randomised trial. Lancet. 1999;353:611-616.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  ALLHAT Officers and Coordinators for the ALLHAT Collaborative Research Group, The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial. Major outcomes in high-risk hypertensive patients randomized to angiotensin-converting enzyme inhibitor or calcium channel blocker vs diuretic: The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). JAMA. 2002;288:2981-2997.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  The SOLVD Investigattors. Effect of enalapril on mortality and the development of heart failure in asymptomatic patients with reduced left ventricular ejection fractions. N Engl J Med. 1992;327:685-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2706]  [Cited by in F6Publishing: 2516]  [Article Influence: 78.6]  [Reference Citation Analysis (0)]
9.  Vermes E, Ducharme A, Bourassa MG, Lessard M, White M, Tardif JC. Enalapril reduces the incidence of diabetes in patients with chronic heart failure: insight from the Studies Of Left Ventricular Dysfunction (SOLVD). Circulation. 2003;107:1291-1296.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  New JP, Bilous RW, Walker M. Insulin sensitivity in hypertensive Type 2 diabetic patients after 1 and 19 days’ treatment with trandolapril. Diabet Med. 2000;17:134-140.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Heise T, Heinemann L, Kristahn K, Berger M, Sawicki PT. Insulin sensitivity in patients with essential hypertension: no influence of the ACE inhibitor enalapril. Horm Metab Res. 1999;31:418-423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
12.  Heinemann L, Heise T, Ampudia J, Sawicki P, Sindelka G, Brunner G, Starke AA. Four week administration of an ACE inhibitor and a cardioselective beta-blocker in healthy volunteers: no influence on insulin sensitivity. Eur J Clin Invest. 1995;25:595-600.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  DREAM Trial Investigators, Bosch J, Yusuf S, Gerstein HC, Pogue J, Sheridan P, Dagenais G, Diaz R, Avezum A, Lanas F. Effect of ramipril on the incidence of diabetes. N Engl J Med. 2006;355:1551-1562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 543]  [Cited by in F6Publishing: 476]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
14.  DREAM On Investigators, Gerstein HC, Mohan V, Avezum A, Bergenstal RM, Chiasson JL, Garrido M, MacKinnon I, Rao PV, Zinman B, Jung H, Joldersma L, Bosch J, Yusuf S. Long-term effect of rosiglitazone and/or ramipril on the incidence of diabetes. Diabetologia. 2011;54:487-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
15.  Abuissa H, Jones PG, Marso SP, O’Keefe JH. Angiotensin-converting enzyme inhibitors or angiotensin receptor blockers for prevention of type 2 diabetes: a meta-analysis of randomized clinical trials. J Am Coll Cardiol. 2005;46:821-826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 325]  [Cited by in F6Publishing: 342]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
16.  Scheen AJ. Renin-angiotensin system inhibition prevents type 2 diabetes mellitus. Part 1. A meta-analysis of randomised clinical trials. Diabetes Metab. 2004;30:487-496.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Dahlöf B, Devereux RB, Kjeldsen SE, Julius S, Beevers G, de Faire U, Fyhrquist F, Ibsen H, Kristiansson K, Lederballe-Pedersen O. Cardiovascular morbidity and mortality in the Losartan Intervention For Endpoint reduction in hypertension study (LIFE): a randomised trial against atenolol. Lancet. 2002;359:995-1003.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3768]  [Cited by in F6Publishing: 3383]  [Article Influence: 153.8]  [Reference Citation Analysis (0)]
18.  Lindholm LH, Persson M, Alaupovic P, Carlberg B, Svensson A, Samuelsson O. Metabolic outcome during 1 year in newly detected hypertensives: results of the Antihypertensive Treatment and Lipid Profile in a North of Sweden Efficacy Evaluation (ALPINE study). J Hypertens. 2003;21:1563-1574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 53]  [Reference Citation Analysis (0)]
19.  Julius S, Kjeldsen SE, Weber M, Brunner HR, Ekman S, Hansson L, Hua T, Laragh J, McInnes GT, Mitchell L. Outcomes in hypertensive patients at high cardiovascular risk treated with regimens based on valsartan or amlodipine: the VALUE randomised trial. Lancet. 2004;363:2022-2031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1920]  [Cited by in F6Publishing: 1757]  [Article Influence: 87.9]  [Reference Citation Analysis (0)]
20.  Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJ, Michelson EL, Olofsson B, Ostergren J, Yusuf S, Pocock S. Effects of candesartan on mortality and morbidity in patients with chronic heart failure: the CHARM-Overall programme. Lancet. 2003;362:759-766.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  McMurray JJ, Holman RR, Haffner SM, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, Buckley BM. Effect of valsartan on the incidence of diabetes and cardiovascular events. N Engl J Med. 2010;362:1477-1490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 434]  [Cited by in F6Publishing: 428]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
22.  Lithell H, Hansson L, Skoog I, Elmfeldt D, Hofman A, Olofsson B, Trenkwalder P, Zanchetti A. The Study on Cognition and Prognosis in the Elderly (SCOPE): principal results of a randomized double-blind intervention trial. J Hypertens. 2003;21:875-886.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
23.  Geng DF, Jin DM, Wu W, Xu Y, Wang JF. Angiotensin receptor blockers for prevention of new-onset type 2 diabetes: a meta-analysis of 59,862 patients. Int J Cardiol. 2012;155:236-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
24.  Cheung BM, Cheung GT, Lauder IJ, Lau CP, Kumana CR. Meta-analysis of large outcome trials of angiotensin receptor blockers in hypertension. J Hum Hypertens. 2006;20:37-43.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Benson SC, Pershadsingh HA, Ho CI, Chittiboyina A, Desai P, Pravenec M, Qi N, Wang J, Avery MA, Kurtz TW. Identification of telmisartan as a unique angiotensin II receptor antagonist with selective PPARgamma-modulating activity. Hypertension. 2004;43:993-1002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 824]  [Cited by in F6Publishing: 836]  [Article Influence: 41.8]  [Reference Citation Analysis (0)]
26.  Schupp M, Janke J, Clasen R, Unger T, Kintscher U. Angiotensin type 1 receptor blockers induce peroxisome proliferator-activated receptor-gamma activity. Circulation. 2004;109:2054-2057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 555]  [Cited by in F6Publishing: 552]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
27.  Rizos CV, Elisaf MS, Liberopoulos EN. Are the pleiotropic effects of telmisartan clinically relevant? Curr Pharm Des. 2009;15:2815-2832.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Nagel JM, Tietz AB, Göke B, Parhofer KG. The effect of telmisartan on glucose and lipid metabolism in nondiabetic, insulin-resistant subjects. Metabolism. 2006;55:1149-1154.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Benndorf RA, Rudolph T, Appel D, Schwedhelm E, Maas R, Schulze F, Silberhorn E, Böger RH. Telmisartan improves insulin sensitivity in nondiabetic patients with essential hypertension. Metabolism. 2006;55:1159-1164.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Usui I, Fujisaka S, Yamazaki K, Takano A, Murakami S, Yamazaki Y, Urakaze M, Hachiya H, Takata M, Senda S. Telmisartan reduced blood pressure and HOMA-IR with increasing plasma leptin level in hypertensive and type 2 diabetic patients. Diabetes Res Clin Pract. 2007;77:210-214.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Negro R, Hassan H. The effects of telmisartan and amlodipine on metabolic parameters and blood pressure in type 2 diabetic, hypertensive patients. J Renin Angiotensin Aldosterone Syst. 2006;7:243-246.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Ichikawa Y. Comparative effects of telmisartan and valsartan on insulin resistance in hypertensive patients with metabolic syndrome. Intern Med. 2007;46:1331-1336.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Vitale C, Mercuro G, Castiglioni C, Cornoldi A, Tulli A, Fini M, Volterrani M, Rosano GM. Metabolic effect of telmisartan and losartan in hypertensive patients with metabolic syndrome. Cardiovasc Diabetol. 2005;4:6.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Sasaki J, Iwashita M, Kono S. Statins: beneficial or adverse for glucose metabolism. J Atheroscler Thromb. 2006;13:123-129.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Collins R, Armitage J, Parish S, Sleigh P, Peto R. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial. Lancet. 2003;361:2005-2016.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Sabatine MS, Wiviott SD, Morrow DA, McCabe CH, Cannon CP. High dose atorvastatin associated with worse glycemic control: a PROVE-IT TIMI 22 substudy [Abstract]. Circulation. 2004;110:S834.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Sever PS, Dahlöf B, Poulter NR, Wedel H, Beevers G, Caulfield M, Collins R, Kjeldsen SE, Kristinsson A, McInnes GT. Prevention of coronary and stroke events with atorvastatin in hypertensive patients who have average or lower-than-average cholesterol concentrations, in the Anglo-Scandinavian Cardiac Outcomes Trial--Lipid Lowering Arm (ASCOT-LLA): a multicentre randomised controlled trial. Lancet. 2003;361:1149-1158.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359:2195-2207.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Rizos CV, Milionis HJ, Kostapanos MS, Florentin M, Kostara CE, Elisaf MS, Liberopoulos EN. Effects of rosuvastatin combined with olmesartan, irbesartan, or telmisartan on indices of glucose metabolism in Greek adults with impaired fasting glucose, hypertension, and mixed hyperlipidemia: a 24-week, randomized, open-label, prospective study. Clin Ther. 2010;32:492-505.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Yusuf S, Teo KK, Pogue J, Dyal L, Copland I, Schumacher H, Dagenais G, Sleight P, Anderson C. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008;358:1547-1559.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Yusuf S, Teo K, Anderson C, Pogue J, Dyal L, Copland I, Schumacher H, Dagenais G, Sleight P. Effects of the angiotensin-receptor blocker telmisartan on cardiovascular events in high-risk patients intolerant to angiotensin-converting enzyme inhibitors: a randomised controlled trial. Lancet. 2008;372:1174-1183.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Yusuf S, Diener HC, Sacco RL, Cotton D, Ounpuu S, Lawton WA, Palesch Y, Martin RH, Albers GW, Bath P. Telmisartan to prevent recurrent stroke and cardiovascular events. N Engl J Med. 2008;359:1225-1237.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Elliott WJ, Meyer PM. Incident diabetes in clinical trials of antihypertensive drugs: a network meta-analysis. Lancet. 2007;369:201-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 762]  [Cited by in F6Publishing: 672]  [Article Influence: 39.5]  [Reference Citation Analysis (0)]
44.  van den Meiracker AH, Jan Danser AH. Aliskiren: the first direct renin inhibitor for hypertension. Curr Cardiol Rep. 2007;9:470-476.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  De Rosa ML, Musella F, Ilardi F, D’Amore C, Luciano R, Maresca F. Effects of antihypertensive therapy on glucose, insulin metabolism, left ventricular diastolic dysfunction and renin system in overweight and obese hypertensives. J Renin Angiotensin Aldosterone Syst. 2013;15:196-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
46.  Uresin Y, Taylor AA, Kilo C, Tschöpe D, Santonastaso M, Ibram G, Fang H, Satlin A. Efficacy and safety of the direct renin inhibitor aliskiren and ramipril alone or in combination in patients with diabetes and hypertension. J Renin Angiotensin Aldosterone Syst. 2007;8:190-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 173]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
47.  Fogari R, Zoppi A, Mugellini A, Lazzari P, Derosa G. Different effects of aliskiren and losartan on fibrinolysis and insulin sensitivity in hypertensive patients with metabolic syndrome. Horm Metab Res. 2010;42:892-896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
48.  Erdös EG. Angiotensin I converting enzyme. Circ Res. 1975;36:247-255.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Yang HY, Erdös EG, Levin Y. A dipeptidyl carboxypeptidase that converts angiotensin I and inactivates bradykinin. Biochim Biophys Acta. 1970;214:374-376.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Uehara M, Kishikawa H, Isami S, Kisanuki K, Ohkubo Y, Miyamura N, Miyata T, Yano T, Shichiri M. Effect on insulin sensitivity of angiotensin converting enzyme inhibitors with or without a sulphydryl group: bradykinin may improve insulin resistance in dogs and humans. Diabetologia. 1994;37:300-307.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Miyata T, Taguchi T, Uehara M, Isami S, Kishikawa H, Kaneko K, Araki E, Shichiri M. Bradykinin potentiates insulin-stimulated glucose uptake and enhances insulin signal through the bradykinin B2 receptor in dog skeletal muscle and rat L6 myoblasts. Eur J Endocrinol. 1998;138:344-352.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Henriksen EJ, Jacob S, Fogt DL, Dietze GJ. Effect of chronic bradykinin administration on insulin action in an animal model of insulin resistance. Am J Physiol. 1998;275:R40-R45.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Huang S, Czech MP. The GLUT4 glucose transporter. Cell Metab. 2007;5:237-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 917]  [Cited by in F6Publishing: 920]  [Article Influence: 54.1]  [Reference Citation Analysis (0)]
54.  Velloso LA, Folli F, Sun XJ, White MF, Saad MJ, Kahn CR. Cross-talk between the insulin and angiotensin signaling systems. Proc Natl Acad Sci USA. 1996;93:12490-12495.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Andreozzi F, Laratta E, Sciacqua A, Perticone F, Sesti G. Angiotensin II impairs the insulin signaling pathway promoting production of nitric oxide by inducing phosphorylation of insulin receptor substrate-1 on Ser312 and Ser616 in human umbilical vein endothelial cells. Circ Res. 2004;94:1211-1218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 157]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
56.  Hoenack C, Roesen P. Inhibition of angiotensin type 1 receptor prevents decline of glucose transporter (GLUT4) in diabetic rat heart. Diabetes. 1996;45 Suppl 1:S82-S87.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Henriksen EJ, Jacob S, Kinnick TR, Teachey MK, Krekler M. Selective angiotensin II receptor antagonism reduces insulin resistance in obese Zucker rats. Hypertension. 2001;38:884-890.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Janke J, Engeli S, Gorzelniak K, Luft FC, Sharma AM. Mature adipocytes inhibit in vitro differentiation of human preadipocytes via angiotensin type 1 receptors. Diabetes. 2002;51:1699-1707.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Sharma AM, Janke J, Gorzelniak K, Engeli S, Luft FC. Angiotensin blockade prevents type 2 diabetes by formation of fat cells. Hypertension. 2002;40:609-611.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Engeli S, Schling P, Gorzelniak K, Boschmann M, Janke J, Ailhaud G, Teboul M, Massiéra F, Sharma AM. The adipose-tissue renin-angiotensin-aldosterone system: role in the metabolic syndrome? Int J Biochem Cell Biol. 2003;35:807-825.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Steinberg HO, Chaker H, Leaming R, Johnson A, Brechtel G, Baron AD. Obesity/insulin resistance is associated with endothelial dysfunction. Implications for the syndrome of insulin resistance. J Clin Invest. 1996;97:2601-2610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1180]  [Cited by in F6Publishing: 1183]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]
62.  Lupi R, Del Guerra S, Bugliani M, Boggi U, Mosca F, Torri S, Del Prato S, Marchetti P. The direct effects of the angiotensin-converting enzyme inhibitors, zofenoprilat and enalaprilat, on isolated human pancreatic islets. Eur J Endocrinol. 2006;154:355-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 70]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
63.  Hall JE, Brands MW, Kivlighn SD, Mizelle HL, Hildebrandt DA, Gaillard CA. Chronic hyperinsulinemia and blood pressure. Interaction with catecholamines? Hypertension. 1990;15:519-527.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Johns DW, Ayers CR, Williams SC. Dilation of forearm blood vessels after angiotensin-converting-enzyme inhibition by captopril in hypertensive patients. Hypertension. 1984;6:545-550.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Kodama J, Katayama S, Tanaka K, Itabashi A, Kawazu S, Ishii J. Effect of captopril on glucose concentration. Possible role of augmented postprandial forearm blood flow. Diabetes Care. 1990;13:1109-1111.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Reaven GM, Lithell H, Landsberg L. Hypertension and associated metabolic abnormalities--the role of insulin resistance and the sympathoadrenal system. N Engl J Med. 1996;334:374-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 968]  [Cited by in F6Publishing: 897]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
67.  De Mattia G, Ferri C, Laurenti O, Cassone-Faldetta M, Piccoli A, Santucci A. Circulating catecholamines and metabolic effects of captopril in NIDDM patients. Diabetes Care. 1996;19:226-230.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Moan A, Risanger T, Eide I, Kjeldsen SE. The effect of angiotensin II receptor blockade on insulin sensitivity and sympathetic nervous system activity in primary hypertension. Blood Press. 1994;3:185-188.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Paolisso G, Scheen A, D’Onofrio F, Lefèbvre P. Magnesium and glucose homeostasis. Diabetologia. 1990;33:511-514.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Paolisso G, Passariello N, Pizza G, Marrazzo G, Giunta R, Sgambato S, Varricchio M, D’Onofrio F. Dietary magnesium supplements improve B-cell response to glucose and arginine in elderly non-insulin dependent diabetic subjects. Acta Endocrinol (Copenh). 1989;121:16-20.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Haenni A, Berglund L, Reneland R, Anderssson PE, Lind L, Lithell H. The alterations in insulin sensitivity during angiotensin converting enzyme inhibitor treatment are related to changes in the calcium/magnesium balance. Am J Hypertens. 1997;10:145-151.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Noto H, Goto A, Tsujimoto T, Noda M. Effect of calcium channel blockers on incidence of diabetes: a meta-analysis. Diabetes Metab Syndr Obes. 2013;6:257-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
73.  Yang Y, Wei RB, Xing Y, Tang L, Zheng XY, Wang ZC, Gao YW, Li MX, Chen XM. A meta-analysis of the effect of angiotensin receptor blockers and calcium channel blockers on blood pressure, glycemia and the HOMA-IR index in non-diabetic patients. Metabolism. 2013;62:1858-1866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
74.  Shen L, Shah BR, Reyes EM, Thomas L, Wojdyla D, Diem P, Leiter LA, Charbonnel B, Mareev V, Horton ES. Role of diuretics, β blockers, and statins in increasing the risk of diabetes in patients with impaired glucose tolerance: reanalysis of data from the NAVIGATOR study. BMJ. 2013;347:f6745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
75.  Levine M, Boyer EW, Pozner CN, Geib AJ, Thomsen T, Mick N, Thomas SH. Assessment of hyperglycemia after calcium channel blocker overdoses involving diltiazem or verapamil. Crit Care Med. 2007;35:2071-2075.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Iwai M, Li HS, Chen R, Shiuchi T, Wu L, Min LJ, Li JM, Tsuda M, Suzuki J, Tomono Y. Calcium channel blocker azelnidipine reduces glucose intolerance in diabetic mice via different mechanism than angiotensin receptor blocker olmesartan. J Pharmacol Exp Ther. 2006;319:1081-1087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
77.  Fukao K, Shimada K, Hiki M, Kiyanagi T, Hirose K, Kume A, Ohsaka H, Matsumori R, Kurata T, Miyazaki T. Effects of calcium channel blockers on glucose tolerance, inflammatory state, and circulating progenitor cells in non-diabetic patients with essential hypertension: a comparative study between azelnidipine and amlodipine on glucose tolerance and endothelial function--a crossover trial (AGENT). Cardiovasc Diabetol. 2011;10:79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
78.  Yamagishi S, Inagaki Y, Nakamura K, Imaizumi T. Azelnidipine, a newly developed long-acting calcium antagonist, inhibits tumor necrosis factor-alpha-induced interleukin-8 expression in endothelial cells through its anti-oxidative properties. J Cardiovasc Pharmacol. 2004;43:724-730.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Popa C, Netea MG, van Riel PL, van der Meer JW, Stalenhoef AF. The role of TNF-alpha in chronic inflammatory conditions, intermediary metabolism, and cardiovascular risk. J Lipid Res. 2007;48:751-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 470]  [Cited by in F6Publishing: 493]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
80.  Nada T, Nomura M, Koshiba K, Kawano T, Mikawa J, Ito S. Clinical study with azelnidipine in patients with essential hypertension. Antiarteriosclerotic and cardiac hypertrophy-inhibitory effects and influence on autonomic nervous activity. Arzneimittelforschung. 2007;57:698-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 10]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
81.  Rizos CV, Elisaf MS. Manidipine: A different dihydropyridine. World J Hypertens. 2011;1:3-6.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Cavalieri L, Cremonesi G. Metabolic effects of manidipine. Am J Cardiovasc Drugs. 2009;9:163-176.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Iimura O, Shimamoto K, Masuda A, Higashiura K, Miyazaki Y, Hirata A, Fukuoka M, Murakami H. Effects of a calcium channel blocker, manidipine, on insulin sensitivity in essential hypertensives. J Diabetes Complications. 1995;9:215-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 18]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
84.  Suzuki S, Ohtomo M, Satoh Y, Kawasaki H, Hirai M, Hirai A, Hirai S, Onoda M, Hinokio Y, Akai H. Effect of manidipine and delapril on insulin sensitivity in type 2 diabetic patients with essential hypertension. Diabetes Res Clin Pract. 1996;33:43-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
85.  Liberopoulos EN, Moutzouri E, Rizos CV, Barkas F, Liamis G, Elisaf MS. Effects of manidipine plus rosuvastatin versus olmesartan plus rosuvastatin on markers of insulin resistance in patients with impaired fasting glucose, hypertension, and mixed dyslipidemia. J Cardiovasc Pharmacol Ther. 2013;18:113-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
86.  Martínez Martín FJ. Manidipine in hypertensive patients with metabolic syndrome: the MARIMBA study. Expert Rev Cardiovasc Ther. 2009;7:863-869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
87.  Fogari R, Zoppi A, Corradi L, Preti P, Malalamani GD, Mugellini A. Effects of different dihydropyridine calcium antagonists on plasma norepinephrine in essential hypertension. J Hypertens. 2000;18:1871-1875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
88.  Pollare T, Lithell H, Mörlin C, Präntare H, Hvarfner A, Ljunghall S. Metabolic effects of diltiazem and atenolol: results from a randomized, double-blind study with parallel groups. J Hypertens. 1989;7:551-559.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Pollare T, Lithell H, Selinus I, Berne C. Sensitivity to insulin during treatment with atenolol and metoprolol: a randomised, double blind study of effects on carbohydrate and lipoprotein metabolism in hypertensive patients. BMJ. 1989;298:1152-1157.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Sheu WH, Swislocki AL, Hoffman B, Chen YD, Reaven GM. Comparison of the effects of atenolol and nifedipine on glucose, insulin, and lipid metabolism in patients with hypertension. Am J Hypertens. 1991;4:199-205.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Lithell H, Pollare T, Vessby B. Metabolic effects of pindolol and propranolol in a double-blind cross-over study in hypertensive patients. Blood Press. 1992;1:92-101.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Taylor EN, Hu FB, Curhan GC. Antihypertensive medications and the risk of incident type 2 diabetes. Diabetes Care. 2006;29:1065-1070.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Bangalore S, Parkar S, Grossman E, Messerli FH. A meta-analysis of 94,492 patients with hypertension treated with beta blockers to determine the risk of new-onset diabetes mellitus. Am J Cardiol. 2007;100:1254-1262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 195]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
94.  Agabiti Rosei E, Rizzoni D. Metabolic profile of nebivolol, a beta-adrenoceptor antagonist with unique characteristics. Drugs. 2007;67:1097-1107.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Messerli FH, Grossman E. beta-Blockers in hypertension: is carvedilol different? Am J Cardiol. 2004;93:7B-12B.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 54]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
96.  Bakris GL, Fonseca V, Katholi RE, McGill JB, Messerli FH, Phillips RA, Raskin P, Wright JT, Oakes R, Lukas MA. Metabolic effects of carvedilol vs metoprolol in patients with type 2 diabetes mellitus and hypertension: a randomized controlled trial. JAMA. 2004;292:2227-2236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 600]  [Cited by in F6Publishing: 509]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
97.  Ayers K, Byrne LM, DeMatteo A, Brown NJ. Differential effects of nebivolol and metoprolol on insulin sensitivity and plasminogen activator inhibitor in the metabolic syndrome. Hypertension. 2012;59:893-898.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 60]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
98.  Lithell H, Pollare T, Berne C, Saltin B. The metabolic and circulatory response to beta-blockade in hypertensive men is correlated to muscle capillary density. Blood Press. 1992;1:20-26.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Lund-Johansen P, Omvik P, Nordrehaug JE. Long-term hemodynamic effects of antihypertensive treatment. Clin Investig. 1992;70 Suppl 1:S58-S64.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Rössner S, Taylor CL, Byington RP, Furberg CD. Long term propranolol treatment and changes in body weight after myocardial infarction. BMJ. 1990;300:902-903.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Ye J. Mechanisms of insulin resistance in obesity. Front Med. 2013;7:14-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 419]  [Cited by in F6Publishing: 450]  [Article Influence: 40.9]  [Reference Citation Analysis (0)]
102.  Agarwal R. Hypertension, hypokalemia, and thiazide-induced diabetes: a 3-way connection. Hypertension. 2008;52:1012-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
103.  Verdecchia P, Reboldi G, Angeli F, Borgioni C, Gattobigio R, Filippucci L, Norgiolini S, Bracco C, Porcellati C. Adverse prognostic significance of new diabetes in treated hypertensive subjects. Hypertension. 2004;43:963-969.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 384]  [Cited by in F6Publishing: 399]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
104.  Barzilay JI, Davis BR, Cutler JA, Pressel SL, Whelton PK, Basile J, Margolis KL, Ong ST, Sadler LS, Summerson J. Fasting glucose levels and incident diabetes mellitus in older nondiabetic adults randomized to receive 3 different classes of antihypertensive treatment: a report from the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). Arch Intern Med. 2006;166:2191-2201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 183]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
105.  Savage PJ, Pressel SL, Curb JD, Schron EB, Applegate WB, Black HR, Cohen J, Davis BR, Frost P, Smith W. Influence of long-term, low-dose, diuretic-based, antihypertensive therapy on glucose, lipid, uric acid, and potassium levels in older men and women with isolated systolic hypertension: The Systolic Hypertension in the Elderly Program. SHEP Cooperative Research Group. Arch Intern Med. 1998;158:741-751.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Kostis JB, Wilson AC, Freudenberger RS, Cosgrove NM, Pressel SL, Davis BR. Long-term effect of diuretic-based therapy on fatal outcomes in subjects with isolated systolic hypertension with and without diabetes. Am J Cardiol. 2005;95:29-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 262]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
107.  Kurtz TW. Chlorthalidone: don’t call it “thiazide-like” anymore. Hypertension. 2010;56:335-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
108.  Woodman R, Brown C, Lockette W. Chlorthalidone decreases platelet aggregation and vascular permeability and promotes angiogenesis. Hypertension. 2010;56:463-470.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Puscas I, Coltau M, Baican M, Pasca R, Domuta G, Hecht A. Vasoconstrictive drugs increase carbonic anhydrase I in vascular smooth muscle while vasodilating drugs reduce the activity of this isozyme by a direct mechanism of action. Drugs Exp Clin Res. 2001;27:53-60.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Siffert W, Fox G, Gros G. The effect of carbonic anhydrase inhibition on the velocity of thrombin-stimulated platelet aggregation under physiological conditions. Biochem Biophys Res Commun. 1984;121:266-270.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Biswas UK, Kumar A. Study on the changes of carbonic anhydrase activity in insulin resistance and the effect of methylglyoxal. J Pak Med Assoc. 2012;62:417-421.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Stears AJ, Woods SH, Watts MM, Burton TJ, Graggaber J, Mir FA, Brown MJ. A double-blind, placebo-controlled, crossover trial comparing the effects of amiloride and hydrochlorothiazide on glucose tolerance in patients with essential hypertension. Hypertension. 2012;59:934-942.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
113.  Preiss D, van Veldhuisen DJ, Sattar N, Krum H, Swedberg K, Shi H, Vincent J, Pocock SJ, Pitt B, Zannad F. Eplerenone and new-onset diabetes in patients with mild heart failure: results from the Eplerenone in Mild Patients Hospitalization and Survival Study in Heart Failure (EMPHASIS-HF). Eur J Heart Fail. 2012;14:909-915.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
114.  Yamaji M, Tsutamoto T, Kawahara C, Nishiyama K, Yamamoto T, Fujii M, Horie M. Effect of eplerenone versus spironolactone on cortisol and hemoglobin A₁(c) levels in patients with chronic heart failure. Am Heart J. 2010;160:915-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
115.  Cutler JA. Thiazide-associated glucose abnormalities: prognosis, etiology, and prevention: is potassium balance the key? Hypertension. 2006;48:198-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
116.  Zillich AJ, Garg J, Basu S, Bakris GL, Carter BL. Thiazide diuretics, potassium, and the development of diabetes: a quantitative review. Hypertension. 2006;48:219-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in F6Publishing: 344]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
117.  Shafi T, Appel LJ, Miller ER, Klag MJ, Parekh RS. Changes in serum potassium mediate thiazide-induced diabetes. Hypertension. 2008;52:1022-1029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 83]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
118.  Smith SM, Anderson SD, Wen S, Gong Y, Turner ST, Cooper-Dehoff RM, Schwartz GL, Bailey K, Chapman A, Hall KL. Lack of correlation between thiazide-induced hyperglycemia and hypokalemia: subgroup analysis of results from the pharmacogenomic evaluation of antihypertensive responses (PEAR) study. Pharmacotherapy. 2009;29:1157-1165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
119.  Kao WH, Folsom AR, Nieto FJ, Mo JP, Watson RL, Brancati FL. Serum and dietary magnesium and the risk for type 2 diabetes mellitus: the Atherosclerosis Risk in Communities Study. Arch Intern Med. 1999;159:2151-2159.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  van Dam RM, Hu FB, Rosenberg L, Krishnan S, Palmer JR. Dietary calcium and magnesium, major food sources, and risk of type 2 diabetes in U.S. black women. Diabetes Care. 2006;29:2238-2243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 145]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
121.  Eriksson JW, Jansson PA, Carlberg B, Hägg A, Kurland L, Svensson MK, Ahlström H, Ström C, Lönn L, Ojbrandt K. Hydrochlorothiazide, but not Candesartan, aggravates insulin resistance and causes visceral and hepatic fat accumulation: the mechanisms for the diabetes preventing effect of Candesartan (MEDICA) Study. Hypertension. 2008;52:1030-1037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 103]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
122.  Jacob S, Klimm HJ, Rett K, Helsberg K, Häring HU, Gödicke J. Effects of moxonidine vs. metoprolol on blood pressure and metabolic control in hypertensive subjects with type 2 diabetes. Exp Clin Endocrinol Diabetes. 2004;112:315-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 24]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
123.  Chazova I, Almazov VA, Shlyakhto E. Moxonidine improves glycaemic control in mildly hypertensive, overweight patients: a comparison with metformin. Diabetes Obes Metab. 2006;8:456-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 52]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
124.  Sanjuliani AF, de Abreu VG, Francischetti EA. Selective imidazoline agonist moxonidine in obese hypertensive patients. Int J Clin Pract. 2006;60:621-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
125.  Chazova I, Schlaich MP. Improved Hypertension Control with the Imidazoline Agonist Moxonidine in a Multinational Metabolic Syndrome Population: Principal Results of the MERSY Study. Int J Hypertens. 2013;2013:541689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
126.  Giordano M, Matsuda M, Sanders L, Canessa ML, DeFronzo RA. Effects of angiotensin-converting enzyme inhibitors, Ca2+ channel antagonists, and alpha-adrenergic blockers on glucose and lipid metabolism in NIDDM patients with hypertension. Diabetes. 1995;44:665-671.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Wright JT, Harris-Haywood S, Pressel S, Barzilay J, Baimbridge C, Bareis CJ, Basile JN, Black HR, Dart R, Gupta AK. Clinical outcomes by race in hypertensive patients with and without the metabolic syndrome: Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). Arch Intern Med. 2008;168:207-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 81]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
128.  Almgren T, Wilhelmsen L, Samuelsson O, Himmelmann A, Rosengren A, Andersson OK. Diabetes in treated hypertension is common and carries a high cardiovascular risk: results from a 28-year follow-up. J Hypertens. 2007;25:1311-1317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 93]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]