Skip to main content
Top

24-10-2014 | Metabolic syndrome | Review | Article

Pharmacological treatment and therapeutic perspectives of metabolic syndrome

Journal: Reviews in Endocrine and Metabolic Disorders

Authors: Soo Lim, Robert H. Eckel

Publisher: Springer US

Abstract

Metabolic syndrome is a disorder based on insulin resistance. Metabolic syndrome is diagnosed by a co-occurrence of three out of five of the following medical conditions: abdominal obesity, elevated blood pressures, elevated glucose, high triglycerides, and low high-density lipoprotein-cholesterol (HDL-C) levels. Clinical implication of metabolic syndrome is that it increases the risk of developing type 2 diabetes and cardiovascular diseases. Prevalence of the metabolic syndrome has increased globally, particularly in the last decade, to the point of being regarded as an epidemic. The prevalence of metabolic syndrome in the USA is estimated to be 34 % of adult population. Moreover, increasing rate of metabolic syndrome in developing countries is dramatic. One can speculate that metabolic syndrome is going to induce huge impact on our lives. The metabolic syndrome cannot be treated with a single agent, since it is a multifaceted health problem. A healthy lifestyle including weight reduction is likely most effective in controlling metabolic syndrome. However, it is difficult to initiate and maintain healthy lifestyles, and in particular, with the recidivism of obesity in most patients who lose weight. Next, pharmacological agents that deal with obesity, diabetes, hypertension, and dyslipidemia can be used singly or in combination: anti-obesity drugs, thiazolidinediones, metformin, statins, fibrates, renin-angiotensin system blockers, glucagon like peptide-1 agonists, sodium glucose transporter-2 inhibitors, and some antiplatelet agents such as cilostazol. These drugs have not only their own pharmacologic targets on individual components of metabolic syndrome but some other properties may prove beneficial, i.e. anti-inflammatory and anti-oxidative. This review will describe pathophysiologic features of metabolic syndrome and pharmacologic agents for the treatment of metabolic syndrome, which are currently available.

1 Introduction

The metabolic syndrome is a constellation of cardiovascular and metabolic risk factors including abdominal obesity, hyperglycemia, dyslipidemia, and high blood pressure which predispose the subject to developing type 2 diabetes and cardiovascular disease (CVD) [1]. The prevalence of metabolic syndrome is increasing worldwide. According to data from the National Health and Nutrition Examination Survey (NHANES) III and NHANESs 1999–2006, the age-adjusted prevalence of metabolic syndrome increased from 29.2 to 34.2 % in the US [2]. More than 40 million US adults seem to be affected by the condition [3]. This increasing trend has been also observed in other regions such as Latin American and Asian countries [46], presenting a major challenge for public health professionals as well as becoming a social and economic problem in the near future.
Since World Health Organization (WHO) proposed a working definition for metabolic syndrome in 1998, several definitions have been proposed for clinical diagnosis of the metabolic syndrome (Table 1). Fundamentally, metabolic syndrome is associated with increased adipose tissue or adipose tissue distribution. In particular, abdominal visceral fat produces more pro-inflammatory cytokines such as interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α) [12, 13], which are linked to insulin resistance and additional biomarkers of systemic inflammation, i.e. hsCRP. Metabolic syndrome in cooperation with low grade inflammation contributes to increased risk of developing diabetes and CVD [14] and furthermore all-cause mortality [15].
Table 1
Criteria proposed for clinical diagnosis of the metabolic syndrome
 
WHO (1998)
AACE (2003)
NCEP-ATPIII (2005)
IDF (2005)
IDF;NHLBI;AHA;WHF;IAS;IASO Harmonizing definition (2009)
Reference
[7]
[8]
[9]
[10]
[11]
Requirement
IGT, IFG, T2D, or lowered insulin sensitivitya plus any 2 of the following
IGT or IFG plus any of the following based on clinical judgment
Any 3 of the following 5 features
Increased WC plus any 2 of the following
Three out of 5 would qualify a person for the metabolic syndrome
Obesity
Men: WHR >0.90; Women: WHR >0.85 and/or BMI >30 kg/m2
BMI ≥25 kg/m2
WC ≥102 cm in men or ≥88 cm in women
Population-specific increased WC
Population- and country-specific WC cutoffsb
Triglycerides (TG)
TG ≥150 mg/dl
TG ≥150 mg/dl
TG ≥150 mg/dl or on TG lowering Rx
TG ≥150 mg/dl or on TG lowering Rx
TG ≥150 mg/dlc
HDL-cholesterol (HDL-C)
HDL-C <40 mg/dl in men or HDL-C <50 mg/dl in women
HDL-C <40 mg/dl in men or HDL-C <50 mg/dl in women
HDL-C <40 mg/dl in men or HDL-C <50 mg/dl in women on HDL-C increasing Rx
HDL-C <40 mg/dl in men or HDL-C <50 mg/dl in women on HDL-C increasing Rx
HDL-C <40 mg/dl in men or HDL-C <50 mg/dl in womenb
Blood pressure
≥140/90 mmHg
≥130/85 mmHg
≥130/85 mmHg or on antihypertensive Rx
≥130/85 mmHg or on antihypertensive Rx
≥130/85 mmHg or on antihypertensive Rx
Glucose
IGT, IFG, or T2D
IGT or lFG (but not diabetes)
≥100 mg/dl (includes diabetes)
≥100 mg/dl (includes diabetes)
≥100 mg/dLd
WHO World Health Organization, NCEP-ATP III National Cholesterol Education Program-Adult Treatment Panel III, AACE American Association of Clinical Endocrinologists, IDF International Diabetes Federation, NHLBI National Heart, Lung, and Blood Institute, AHA American Heart Association, WHF World Heart Federation, IAS International Atherosclerosis Society, IASO International Association for the Study of Obesity, T2D type 2 diabetes, IGT impaired glucose tolerance, IFG impaired fasting glucose, WHR waist-to-hip ratio, WC waist circumference, BMI body mass index, TG triglycerides, HDL-C HDL-cholesterol, Rx therapy
aInsulin sensitivity measured under hyperinsulinemic-euglycemic conditions
bThe IDF cut points are recommended for non-Europeans and either the IDF or AHA/NHLBI cut points used for people of European origin until more data are available
cThe most commonly used drugs for elevated triglycerides and reduced HDL-C are fibrates and nicotinic acid. A patient taking 1 of these drugs can be presumed to have high triglycerides and low HDL-C. High-dose ω-3 fatty acids presumes high triglycerides
dMost patients with type 2 diabetes mellitus will have the metabolic syndrome by the proposed criteria
Since the impact of metabolic syndrome on health is enormous, much effort has been made to find optimal therapeutic agents. Currently, the best therapeutic approach for the metabolic syndrome is lifestyle modification including weight loss. However, pharmacological treatments are often needed in people with metabolic syndrome, due to the great difficulty for patients to take measures to change their lifestyle permanently. In people having diseases related to metabolic syndrome, a more broad-based pharmacological agent that modifies insulin resistance may be a good initial choice, i.e., thiazolidinedione (TZD) for diabetic patients with metabolic syndrome. Based on this concept, the treatment of the metabolic syndrome can be approached from very different angles, since each individual may have a different multifaceted health problem (Fig. 1).
In this review, we introduce pathophysiologic features of metabolic syndrome briefly and discuss the potential role of pharmacological agents that can be used focusing on specific component(s).

2 Pathophysiologic features of metabolic syndrome

2.1 Insulin resistance

The major underlying pathophysiology of the metabolic syndrome is insulin resistance. Insulin resistance reflects defects in insulin action in insulin sensitive organs, i.e. adipose tissue, skeletal/cardiac muscle and liver. In adipose tissue, this is a manifested initially as defects in the anti-lipolytic effects of insulin whereas in muscle the defect is insulin-mediated glucose uptake with subsequent reductions in glycogen biosynthesis. This results from inhibition of insulin-stimulated insulin-receptor substrate (IRS)-1 tyrosine phosphorylation and eventually reduced IRS-1–associated phosphatidyl inositol 3 kinase (PI3K) activity [16]. In liver, insulin resistance is manifested by defects in the ability of insulin to suppress glucose production, a defect that relates to maintenance of gluconeogenic enzymes and increased gluconeogenic substrate delivery.
Thus, insulin resistance indicates a pathophysiological condition in which a normal insulin concentration does not adequately produce a normal insulin response in the peripheral target tissues. In this metabolic setting, pancreatic β-cells secrete more insulin, in part to overcome the hyperglycemia and hyperinsulinaemia results. Although hyperinsulinaemia may in part compensate for some insulin resistance, it may contribute to others, i.e. salt retention, hypertension, and hepatic steatosis [17]. An inability of β-cells over time to produce sufficient insulin leads to impaired fasting glucose, impaired glucose tolerance and ultimately type 2 diabetes mellitus [16].
Insulin resistance commonly accompanies abdominal obesity. Although insulin-resistant individuals do not need to be obese, they almost always have preferential fat accumulation in the visceral depot. This is of particular interest in that ordinarily adipocytes obtained from visceral adipose tissue are much less insulin responsive than adipocytes obtained from subcutaneous depots, and additionally abdominally obese subjects also demonstrate more insulin resistance than subjects in whom excess body fat distribution is less central [18]. Some subjects who are not obese may also have insulin resistance associated with ectopic fat accumulation in metabolically active tissues such as liver and muscle [19].

2.2 Pro-inflammatory state, oxidative stress, and renin-angiotensin system in metabolic syndrome

Metabolic syndrome is also considered to be a low-grade pro-inflammatory/pro-oxidative state [12, 20]. The multicenter Insulin Resistance Atherosclerosis Study had shown a linear relationship between the inflammatory marker high-sensitivity C-reactive protein (hsCRP) and the number of metabolic derangements [20]. The large amount of adipose tissue induces systemic inflammation due to an increase of secretory factors derived from adipocytes and macrophages embedded in this tissue [21]. In a population-based non-diabetic Swedish cohort, lipoprotein-associated phospholipase A2 was associated with metabolic syndrome, and each component was related to increased risk for incident CVD in a dose-dependent manner [22]. Thus, hsCRP is the most well-known biomarker for inflammation related to cardiovascular and metabolic risk [23, 24]. Other inflammatory markers such as fibrinogen [25], apolipoprotein-B [26], uric acid [27], and adhesion molecules [28] are also known to be associated with metabolic syndrome.
Atherogenic dyslipidemia manifested by high plasma triglycerides and low HDL-C levels, a strong marker for future cardiovascular events, is also associated with inflammation and oxidative stress in the metabolic syndrome [29]. HDL function is also affected by pro-inflammatory cytokines and oxidative stress [30]. The activation of the renin-angiotensin system (RAS) further promotes inflammation, lipogenesis and reactive oxygen species generation and impairs insulin signaling [31]. Consequently, the increase in circulating RAS, for which adipose tissue is partially responsible, represents a link between high blood pressure, insulin resistance, and inflammation.

3 Pharmacological treatment of metabolic syndrome

In addition to lifestyle including weight loss, a targeted approach for control of individual components of the metabolic syndrome is often necessary. Because there are drugs proven effective in reducing specific components of metabolic syndrome, optimal pharmacological management must be individualized.
First, because weight loss in patients with the metabolic syndrome increases insulin sensitivity [32], anti-obesity drugs should be considered. In the USA the list includes, phentermine, extended release phentermine/topiratmate, lorcaserin, orlistat, and now sustained release bupropion/naltrexone and the glucagon like peptide-1 (GLP-1) agonist liraglutide. Drugs such as TZD or metformin, which are insulin sensitizers, are recommended for insulin resistant patients. Metformin, rosiglitazone and pioglitazone are known to prevent type 2 diabetes in patients with and without the metabolic syndrome [3335] and pioglitazone also increases HDL-C and reduces triglycerides and hepatic steatosis [36]. Lipid-lowering agents such as statins are drugs of choice for atherogenic dyslipidemia [37]. Most but not all metabolic syndrome patients will be statin-eligible; moreover fibrates may reduce CVD events in patients who are hypertriglyceridemic and have reductions in HDL-C [38]. New antidiabetic agents such as GLP-1 agonists and sodium glucose transporter-2 (SGLT-2) inhibitors result in some weight loss and are candidate drugs particularly in metabolic syndrome patients with or at risk for diabetes [39, 40]. RAS blockers are effective in decreasing blood pressure and other cardiovascular risk [41]. Cilostazol, an antiplatelet agent, is also known to improve atherogenic dyslipidemia and increase nitric oxide levels [42]. The mechanism of action, targeted components of metabolic syndrome, additional benefits, and possible side effect of each drug are shown in Table 2.
Table 2
Metabolic syndrome drugs and therapeutic targets in metabolic syndrome, and their side effect
Medications
Mode of action
Main target of metabolic syndrome
Additional benefit
Side effect
Ref.
Lorcaserin
Selective 5-HT 2C agonist
(Abdominal) obesity
TG ↓
Attention deficit or memory problem
[43]
LDL-C ↓
Extended release phentermine + topiramate combination
Voltage-dependent sodium channels, glutamate receptors, and carbonic anhydrase, and augments the activity of γ-aminobutyrate
(Abdominal) obesity
Insulin sensitivity ↑
Pulse rate ↑
[44]
Sustained release bupropion and naltrexone
Alterations in the hypothalamic melanocortin system and brain reward systems that influence food craving and mood
(Abdominal) obesity
Insulin sensitivity ↑
Nausea
[45]
Pulse rate ↑
Blood pressure ↑
Thiazolidinedione
Peroxisome proliferator-activated receptor-γ agonist
High glucose
TG ↓
Fluid retention or weight gain
[46]
HDL-C ↑
Bone mineral density ↓
Inflammation ↓
 
Metformin
Increase in AMPK activity
High glucose
Inflammation ↓
Gastrointestinal trouble
 
Lactic acidosis
Statins
HMG-CoA reductase inhibitor
High TG
LDL-C ↓
Glucose ↑
[37]
Inflammation ↓
Muscle weakness
Fibrates
Peroxisome proliferator-activated receptor-α agonist
High TG
Inflammation ↓
Muscle side effect combined with statin
[47]
Low HDL-C
ACE inhibitors/ARBs
Renin-angiotensin system blockers
High blood pressure
Inflammation ↓
Cough with ACE inhibitor
[48]
Insulin sensitivity ↑
GLP-1 agonists
Stimulation of GLP-1 receptor
High glucose
Body weight ↓
Gastrointestinal trouble
[49]
Blood pressure ↓
SGLT-2 inhibitors
Inhibition of glucose reabsorption in sodium glucose transporter-2
High glucose
Body weight ↓
Urinary tract and genital infection
[50]
Blood pressure ↓
Cilostazol
PDE3 inhibitor
TG ↓
Bleeding tendency
[42]
HDL-C ↑
Inflammation ↓
AMPK AMP-activated protein kinase, HMG-CoA 3-hydroxy-3-methylglutaryl-coenzyme A, ACE angiotensin converting enzyme, ARB angiotensin receptor blocker, GLP-1 glucagon-like peptide-1, SGLT-2 sodium glucose transporter-2, PDE3 phosphodiesterase 3, Ref. reference

3.1 Anti-obesity drugs

Previously, anti-obesity drugs such as sibutramine and orlistat had shown that they reduced body weight and central obesity, resulting in improvement of the components of metabolic syndrome [51]. Rimonabant improved glycemic homeostasis and lipid components of metabolic syndrome: increase in HDL-C levels by 23 % and decrease in triglyceride levels by 15 % [52, 53]. However, due to adverse effects, many anti-obesity drugs, i.e. sibutramine and rimonabant were withdrawn from the market [54]. Phentermine and orlistat remain available; however, several new drugs are now available in the USA and increasingly around the world to consider.

3.1.1 Lorcaserin

Lorcaserin (Belviq®) is approved for its use in obese adults who have high blood pressure, high cholesterol, or type 2 diabetes. In therapeutic doses, lorcaserin acts as a selective 5-HT 2C agonist on pro-opiomelanocortin neurons, which in turn causes release of α-melanocyte-stimulating hormone (α-MSH). Further α-MSH acts on melanocortin 4 receptors in the paraventricular nucleus in the hypothalamus, leading to a decrease in appetite [55]. A multicenter, double-blinded study showed 47.5 % in lorcaserin group and 20.3 % in placebo group lost 5 % or more of body weight after 1 year treatment [43]. At the end of the second year weight loss was maintained by 67.9 % patients who continued to receive lorcaserin, whereas 50.3 % could maintain weight loss who received placebo [43]. Plasma triglycerides and LDL-C levels were significantly lower in the lorcaserin group. These data support that lorcaserin might be beneficial in obese metabolic syndrome subjects with atherogenic dyslipidemia. However, it has been reported that lorcaserin may cause attention deficits and cognitive impairment [43, 55]. Other common side effects include headache, dizziness, fatigue, nausea, dry mouth, and constipation [55].

3.1.2 Extended release phentermine and topiramate combination

A combination of extended release phentermine and topiramate (Qysmia®) has also been recently approved by the US FDA. Phentermine works by reducing appetite and topiramate by unclear mechanism(s) [56]. Topiramate blocks voltage-dependent sodium channels, glutamate receptors, and carbonic anhydrase, and augments the activity of γ-aminobutyrate [57]. The combination of extended release phentermine and topiramate is well tolerated and can result in impressive weight reduction despite low doses of each drug that would have minimal effects alone. Target subjects for the drug are obese or overweight people who suffer from conditions such as diabetes, hypertension and/or hypercholesterolemia. Thus, patients with metabolic syndrome would be good candidates. However, there is still a safety concern with this combination such as elevation in blood pressure and pulse rate for phentermine and peripheral neuropathy and possible teratogenic effects for topiramate [58].

3.1.3 Sustained release bupropion and naltrexone

The US FDA recently approved sustained release bupropion and naltrexone (Contrave®) in September, 2014. In part this decision was based on the results of CONTRAVE Obesity Research-II (COR-II), a double-blind, placebo-controlled study of 1,496 obese (BMI 30–45 kg/m2) or overweight (27–45 kg/m2) patients with dyslipidemia and/or hypertension [45]. Treatment of combined naltrexone sustained-release (SR) (32 mg/day) plus bupropion SR (360 mg/day) (NB32) for up to 56 weeks resulted in a greater weight loss than placebo at week 28 (−6.5 % vs. −1.9 %) and week 56 (−6.4 % vs. −1.2 %) (both p < 0.001). In addition, the combination produced greater improvements in various cardiometabolic risk markers. The most common adverse event was nausea, which was generally mild and transient. Although the mechanism of action of the combination of sustained release bupropion and naltrexone for weight reduction has not been fully revealed, the combination may induce alterations in the hypothalamic melanocortin system as well as brain reward systems that influence food craving and mood [59].

3.2 Insulin sensitizers

3.2.1 Thiazolidinedione (TZD)

TZD is a synthetic ligand of peroxisome proliferator-activated receptor-γ (PPARγ), which is a member of the nuclear hormone receptor family and contributes to many biological processes such as glucose regulation and lipid metabolism [46]. PPARγ is known to regulate expression of the gene encoding prodifferentiation transcription-factor pancreas duodenum homeobox-1 (PDX-1) in pancreatic β-cells [60], which regulates glucose-responsive insulin gene transcription as well as the transcription of glucose transporter-2 and glucokinase [61, 62]. Many studies demonstrate that TZDs reduce insulin resistance, a core defect in metabolic syndrome, via several mechanisms [63]. First, TZDs reduces the intracellular levels of toxic lipid metabolites, resulting in less lipotoxicity [64]. TZDs also protect against the cytostatic effect of free fatty acids and restores glucose-mediated insulin release [65]. TZDs also increase insulin sensitivity in the liver and muscle tissue [66].
In the Actos Now for Prevention of Diabetes (ACT NOW) study, pioglitazone decreased the rate of conversion of subjects with impaired glucose tolerance to type 2 diabetes by 72 % and significantly improved the disposition index [35]. In the A Diabetes Outcome Progression Trial (ADOPT) study, rosiglitazone treatment improved insulin sensitivity and prevented decline of β-cell function more than either metformin or sulfonylurea [67]. Since TZDs, pioglitazone more than rosiglitazone, improve various features of metabolic syndrome such as hyperglycemia, elevated blood pressure, hypertriglyceridemia, and low HDL-C, TZDs are an useful agent for the treatment of the metabolic syndrome [36, 68].
TZDs also decrease inflammatory factors such as hsCRP and promote adipose tissue differentiation in subcutaneous adipose tissue regions, which increases the synthesis of adiponectin, thereby further reducing insulin resistance. TZDs are also known to reduce circulating levels of free fatty acids, resistin, IL-6, TNF-α, and ICAM-1 [69, 70]. However, side effects of the TZDs such as fluid retention and weight gain are important to consider. In addition, adverse effects of TZDs on bone health and perhaps bladder cancer, although not yet confirmed, prevent its wide use in clinical practice [7173].

3.2.2 Metformin

Metformin increases AMP-activated protein kinase (AMPK) activity, a pharmacological target for the treatment of insulin resistance. Metformin is a biguanide and is recommended as a first line drug for type 2 diabetes by the American Diabetes Association and European Association for the Study of Diabetes guidelines [74]. Recent evidence indicates that metformin can also have antidiabetic activity through mediation of a family of proteins known as sirtuins, a type of histone deacetylases controlling cellular metabolism through regulation of the expression of several genes [75]. Metformin also has modest anti-inflammatory properties [76]. Because metformin like TZDs reduces the incidence of new onset diabetes in individuals with metabolic syndrome, it should be seriously considered in metabolic syndrome subjects with impaired fasting glucose or impaired glucose tolerance (prediabetes).

3.3 Lipid lowering agents

Statin, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor, and fibrate, a PPARα agonist, are good candidates for pharmacologic agents for metabolic syndrome with dyslipidemia. A combination of statins and fibrates can be recommended to control atherogenic dyslipidemia in metabolic syndrome.

3.3.1 Statins

Statins are competitive inhibitors of HMG-CoA reductase, blocking the rate-limiting step in cholesterol biosynthesis [77]. Hence, statins are effective in reducing circulating total and LDL-C levels. Of interest, however, is that the lowering of LDL-C is not via a reduction in LDL-C synthesis that occurs via lipoprotein lipase-mediated hydrolysis of VLDL, but by reducing the intrahepatic cholesterol pool size and increasing LDL receptors [78, 79].
Much evidence has proven that statin therapy significantly and substantially reduces cardiovascular risk [79]. Many prospective, randomized trials such as the COmparative study with rosuvastatin in subjects with METabolic Syndrome (COMETS) and the Measuring Effective Reductions in Cholesterol Using Rosuvastatin TherapY I (MERCURY I) trial showed that statins improved atherogenic dyslipidemia in people with metabolic syndrome [8082]. In a meta-analysis from 27 randomised trials, statin therapy in people at low risk of vascular disease was also effective in reducing CVD [83]. Although the use of statins for the primary prevention in subjects with a low CVD risk remains suspensive in its cost-effectiveness [84], statin treatment has demonstrated consistently a significant reduction in cardiovascular and all-cause mortality in people at high risk of CVD [37, 85, 86].
Independently of their action upon cholesterol; statins diminish oxidative stress and improve endothelial function [87]. Statins also have anti-inflammatory properties by inhibition of isoprenoid (farnesyl or geranyl) formation and blockage of the activation of the Rho and Rac pathways which are involved in the activation of Nuclear Factor(NF)-κB as well as direct inhibition of the NF-κB activation [88]. With these beneficial effects on other cardiovascular risk as well as lipid-lowering property, statins are recommended as the cornerstone of treatment of dyslipidemia in most patients with metabolic syndrome. The risk estimator developed by the American College of Cardiology/American Heart Association Task Force on Practice Guidelines can be utilized to estimate risk in patients with the metabolic syndrome without known CVD [89].
Recently, several meta-analyses of randomized controlled trials have suggested more insulin resistance following statin treatment [90, 91]. The mechanisms by which statins aggravate insulin resistance have not been fully elucidated. However, statins may decrease the production of metabolites of HMG-CoA, such as isoprenoids, which thereupon down regulate insulin-responsive glucose transporter (GLUT)-4, thereby decreasing glucose uptake [92]. This pharmacological effect has been documented for simvastatin and atorvastatin [92, 93]. In patients with prediabetes, this mechanism could be important in the transition from prediabetes to diabetes [94]. Another mechanism could be the recently described weight gain seen in statin-treated patients [95]. It would be prudent that the risk of deteriorating insulin sensitivity and weight gain in statin-treated patients be balanced with the more substantial benefits in reducing cardiovascular risks [96].

3.3.2 Fibrates

Fibrates are PPARα activators and useful for the treatment of dyslipidemia, particularly for patients with hypertriglyceridemia and low levels of HDL-C [47]. Fibrates also diminish circulating levels of fibrinogen, IL-1, IL-6, and hsCRP, leading to reduction of NF-κB and improvement of vascular function [47]. In the FEILD study, fenofibrate treatment reduced cardiovascular outcomes by 27 % in subjects with hypertriglyceridemia and low HDL-cholesterol, although it did not significantly reduce the risk of the primary outcome of coronary events [97]. Most studies have shown that fibrates reduce triglycerides levels by 30–45 % [98, 99] and increase HDL-C by up to 25 % but most often by <10 % [100]. Fenofibrate also decreased fibrinolysis inhibitor concentrations and improved endothelial function in metabolic syndrome patients, suggesting another potential mechanism for protection against CVD [101].
In some patients, the treatment for atherogenic dyslipidemia requires two drugs with different targets. Statin and fibrate combination is a good combination for treatment of lipid components and CVD risk in metabolic syndrome patients [102].

3.4 RAS blockers

There is an important relationship of RAS with insulin resistance and endothelial dysfunction, which are found in people with metabolic syndrome and obesity-related hypertension [103, 104]. Angiotensin II inhibits insulin signaling and produces oxidative stress that accelerates hyperglycemia and atherosclerosis [105]. Therefore, blockers of the RAS are an important component of therapeutic agents for metabolic syndrome. There are two types of RAS blockers: angiotensin converting enzyme (ACE) inhibitor and angiotensin-II receptor blocker (ARB).
Enalapril, an ACE inhibitor, is hydrolyzed to the active form enalaprilat, which reduces the plasma levels of angiotensin-II, leading to a decrease in blood pressure by peripheral vasodilatation [48]. Ramipril, another ACE inhibitor, proved cardiovascular benefit in a large clinical trial [106]. A fixed-dose combination of perindopril with indapamide showed substantial protection against cardiovascular morbidity and mortality [107]. ARBs are also known to exert blood pressure lowering by reducing the secretion of vasopressin and aldosterone [108].
Angiotensin-II also has a central role in glucose metabolism that includes activation of insulin-stimulated mitogenic pathways that promote vascular smooth muscle proliferation via mitogen-activated protein kinase (MAPK), but suppression of pathways involved in glucose transport such as PI-3 K [109]. The beneficial effect of ACE inhibitors on glucose metabolism is demonstrated by clinical trials such as the HOPE (Heart Outcomes Prevention Evaluation) Study, which showed a reduced rate of new onset diabetes mellitus in patients taking the ACE inhibitor ramipril [110] although this effect has been variable [111]. ARBs also have insulin sensitizing and antidiabetic effects, induce PPARγ activity and reduce serum uric acid levels [112].
ACE inhibitors also have anti-inflammatory effects, pleiotropic actions of the drugs that are not related with ACE itself. ACE inhibitors increase the synthesis of adiponectin [113]. Furthermore, ACE inhibitors reduce oxidative stress and endothelial dysfunction particularly in subjects with depressed cardiac function [114]. ARBs also have anti-inflammatory effects: reduction of circulating levels of IL-6 and TNF-α [115] and decrease in hsCRP and ICAM-1 levels [116]. ARBs have good tolerability and safety profile, which have been reported to be similar to placebo [117]. A recent analysis with Cardiovascular Health Study showed that RAS blocking agents reduced cardiovascular events in patients with metabolic syndrome [118]. Thus, RAS blockers are indicated for the treatment of elevated blood pressure in people with metabolic syndrome.

3.5 Novel antidiabetic agents

3.5.1 Glucagon like peptide-1 (GLP-1) agonists

GLP-1 agonists are an incretin-based therapy, which is derived from the concept that ingested glucose results in a greater increase in insulin secretion compared with intravenous glucose administration. GLP-1 and glucose-dependent insulinotropic polypeptide (GIP), two intestinal-derived hormones, stimulate insulin secretion in pancreatic β-cells [49]. In addition, GLP-1 infusion has been shown to inhibit glucagon secretion, slow gastric emptying, and reduce food intake [119, 120]. Endogenous GLP-1 is rapidly inactivated by dipeptidyl peptidase-4 enzyme. To overcome this rapid inactivation of native GLP-1, synthetic GLP-1 receptor agonists with prolonged action profiles have been developed for treatment of patients with type 2 diabetes [121].
GLP-1 agonists improve hyperglycemia by enhancing glucose-stimulated insulin secretion through activation of cyclic adenosine monophosphate (cAMP). Increased cAMP upregulates protein kinase A (PKA) and exchange protein activated by cAMP, which leads to rapid increases in intracellular calcium and insulin exocytosis in a glucose-dependent manner [122, 123]. Moreover, GLP-1 agonists have the potential to reduce insulin resistance which helps reverse insulin resistance-associated defects in the failing β-cells. GLP-1 is also known to attenuate endoplasmic reticulum stress via activation of PKA [124].
Specifically, among GLP-1 agonists, liraglutide 3 mg (Saxenda®) has been approved by the US FDA as an antiobesity drug. In addition to stimulation of insulin release and decrease of glucagon secretion in response to hyperglycemia, liraglutide at higher doses, i.e. 3 mg, has resulted in more weight loss presumably by greater reductions in appetite and energy intake. In a large scale, randomized clinical trial, treatment with liraglutide 3 mg induced sustained weight loss by 7.2 kg over 20 weeks, reduced blood pressure, and improved glycemic control [125]. Despite the higher dose, liraglutide was reasonably well tolerated with gastrointestinal side effects being most commonly encountered. Thus, liraglutide 3 mg provides a sustainable weight loss and improvement of several metabolic syndrome-related comorbidities. In summary, GLP-1 agonists seem to be a good candidate drug class for metabolic syndrome subjects with hyperglycemia and overweight/obesity since it enhances insulin sensitivity and reduces body weight as well as increases insulin secretory function.

3.5.2 Sodium glucose transporter-2 (SGLT-2) inhibitors

The kidney plays an important role in glucose homeostasis by mediating reabsorption of glucose from the glomerular filtrate, which happens through SGLT-2 and SGLT-1 [50]. SGLT-2 inhibitors block 90 % of reabsorption of filtered glucose in the proximal tubules, leading to increased urinary glucose excretion, which eventually reduces hyperglycemia [126]. In 2013–2014, the US FDA approved three drugs in this class: canagliflozin, empagliflozin, and dapagliflozin as antidiabetic medications. In addition to glucose lowering efficacy [127, 128], SGLT-2 inhibitors result in loss of calories and weight loss [129]. In fact, treatment with dapagliflozin and metformin reduced body weight by 5 % after 24 weeks [40]. Initial weight loss may come from fluid loss, as urinary glucose excretion results in mild osmotic diuresis. But dapagliflozin treatment reduced body weight, predominantly by reducing fat mass [40]. This finding suggests that caloric loss from glycosuria, not fluid loss, by SGLT-2 inhibition is mainly responsible for the decrease in body weight and fat mass. SGLT-2 inhibitors also reduced blood pressures by 3–9 mmHg in clinical studies [130, 131], which is likely mediated by osmotic diuresis.
SGLT-2 inhibitors have additional benefits in management of metabolic syndrome. Treatment with empagliflozin, another SGLT-2 inhibitor, showed anti-inflammatory effect with a reduction of IL-6, TNF-α, and monocyte chemotactic protein-1 levels in an animal study [132]. SGLT-2 inhibitors also affect lipid metabolism. Dapagliflozin treatment increased HDL-C and reduced triglycerides [133, 134]. In contrast, canagliflozin treatment increased HDL-C and LDL-C, and resulted in inconsistent changes in triglycerides [135]. SGLT-2 inhibitors also reduced serum uric acid levels by approximately 1 mg/dL [127, 131]. Hyperuricemia is related to insulin resistance and some consider this a component of metabolic syndrome.
Based on these data, SGLT-2 inhibitors may have the potential to reduce cardiovascular risk not only via glucose lowering effect but via beneficial effects on body weight, blood pressure and serum uric acid in patients with metabolic syndrome [136].

3.6 Cilostazol

Among several antiplatelet agents, cilostazol has properties that would be helpful for metabolic syndrome subjects. Cilostazol, a selective phosphodiesterase-3 (PDE-3) inhibitor, is an antithrombotic agent with vasodilating properties. Cilostazol activates AMPK and causes phosphorylation of endothelial nitric oxide synthase (eNOS), leading to increased production of nitric oxide (NO), while it inhibits cytokine-induced NF-kB activation and suppresses VCAM-1 gene expression [137]. Cilostazol attenuates cytokine-induced expression of the iNOS gene by inhibiting NF-kB following AMPK activation in vascular smooth muscle cell [138]. Cilostazol also appears to exert a beneficial effect against hepatic steatosis by suppressing mitogen-activated protein kinase (MAPK) activation induced by oxidative stress [139]. In a clinical trial, cilostazol reduced triglycerides by 15.8 % and increased HDL-C by 12.8 % [42]. Increases in lipoprotein lipase activity by cilostazol treatment seems to be responsible for these lipid changes [140]. Thus, cilostazol may be a candidate drug for metabolic syndrome subjects at high risk of atherosclerosis.

4 Conclusion

Metabolic syndrome is a pleiotropic pathophysiology related to increasing body fat and/or distribution and insulin resistance. Metabolic syndrome is also a public health problem predicting higher rates of type 2 diabetes and CVD. Metabolic syndrome is also a pro-inflammatory condition that involves vascular endothelium and adipose tissue. Adipose tissue secretes many bioactive substances (known as adipocytokines) and inflamed fat contributes to development and complications of metabolic syndrome [141, 142].
Recently approved anti-obesity drugs can be prescribed to reduce body weight, particularly abdominal visceral fat. Other medications such as TZDs, metformin, lipid-lowering medications, RAS blockers, and cilostazol exert many metabolic effects including anti-inflammation.
In this review, we have discussed potential therapeutic agents that can help reduce insulin resistance fundamentally and others that target specific components of metabolic syndrome (Table 2).

Conflicts of Interest

None.
CVD
cardiovascular disease
HDL-C
high-density lipoprotein cholesterol
α-MSH
S-melanocyte-stimulating hormone
GLP-1
glucagon like peptide-1
SGLT-2
sodium glucose transporter-2
hsCRP
high sensitivity C-reactive protein
IL-6
interleukin 6
TNF-α
tumor necrosis factor-α
IRS
insulin-receptor substrate
FDA
Food and Drug Association
TZD
Thiazolidinedione
PPARγ
peroxisome proliferator-activated receptor-γ
PDX-1
pancreas duodenum homeobox-1
HMG-CoA
3-hydroxy-3- methylglutaryl- coenzyme A
ACE
angiotensin-converting enzyme
ARBs
angiotensin II receptor blockers
ICAM
intracellular adhesion molecule
VCAM
vascular cell adhesion molecule
cAMP
cyclic adenosine monophosphate
PKA
protein kinase A
MAPK
mitogen-activated protein kinase
eNOS
endothelial nitric oxide synthase
NO
nitric oxide
Literature
1.
Grundy SM, Brewer Jr HB, Cleeman JI, Smith Jr SC, Lenfant C. Definition of metabolic syndrome: Report of the National Heart, Lung, and Blood Institute/American Heart Association conference on scientific issues related to definition. Circulation. 2004;109(3):433–8.PubMed
2.
Mozumdar A, Liguori G. Persistent increase of prevalence of metabolic syndrome among U.S. adults: NHANES III to NHANES 1999–2006. Diabetes Care. 2011;34(1):216–9.PubMedCentralPubMed
3.
Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: Findings from the third National Health and Nutrition Examination Survey. JAMA. 2002;287(3):356–9.PubMed
4.
Nestel P, Lyu R, Low LP, et al. Metabolic syndrome: Recent prevalence in East and Southeast Asian populations. Asia Pac J Clin Nutr. 2007;16(2):362–7.PubMed
5.
Lim S, Shin H, Song JH, et al. Increasing prevalence of metabolic syndrome in Korea: The Korean National Health and Nutrition Examination Survey for 1998–2007. Diabetes Care. 2011;34(6):1323–8.PubMedCentralPubMed
6.
Rojas R, Guilar-Salinas CA, Jimenez-Corona A, et al. Metabolic syndrome in Mexican adults: Results from the National Health and Nutrition Survey 2006. Salud Publica Mex. 2010;52 Suppl 1:S11–8.PubMed
7.
Alberti KG, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: Diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med. 1998;15(7):539–53.PubMed
8.
Bloomgarden ZT. American Association of Clinical Endocrinologists (AACE) consensus conference on the insulin resistance syndrome: 25–26 August 2002, Washington, DC. Diabetes Care. 2003;26(4):1297–303.PubMed
9.
Grundy SM, Cleeman JI, Daniels SR, et al. Diagnosis and management of the metabolic syndrome: An American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation. 2005;112(17):2735–52.PubMed
10.
Alberti KG, Zimmet P, Shaw J. The metabolic syndrome–a new worldwide definition. Lancet. 2005;366(9491):1059–62.PubMed
11.
Alberti KG, Eckel RH, Grundy SM, et al. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and international association for the Study of Obesity. Circulation. 2009;120(16):1640–5.PubMed
12.
Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the link between insulin resistance, obesity and diabetes. Trends Immunol. 2004;25(1):4–7.PubMed
13.
Morley JE. The metabolic syndrome and aging. J Gerontol A Biol Sci Med Sci. 2004;59(2):139–42.PubMed
14.
Haffner SM, Valdez RA, Hazuda HP, et al. Prospective analysis of the insulin-resistance syndrome (syndrome X). Diabetes. 1992;41(6):715–22.PubMed
15.
Isomaa B, Almgren P, Tuomi T, et al. Cardiovascular morbidity and mortality associated with the metabolic syndrome. Diabetes Care. 2001;24(4):683–9.PubMed
16.
Petersen KF, Shulman GI. Etiology of insulin resistance. Am J Med. 2006;119(5 Suppl 1):S10–6.PubMedCentralPubMed
17.
Gill H, Mugo M, Whaley-Connell A, Stump C, Sowers JR. The key role of insulin resistance in the cardiometabolic syndrome. Am J Med Sci. 2005;330(6):290–4.PubMed
18.
Jensen MD, Haymond MW, Rizza RA, Cryer PE, Miles JM. Influence of body fat distribution on free fatty acid metabolism in obesity. J Clin Invest. 1989;83(4):1168–73.PubMedCentralPubMed
19.
Lim S, Meigs JB. Links between ectopic fat and vascular disease in humans. Arterioscler Thromb Vasc Biol. 2014;34(9):1820–6.PubMed
20.
Festa A, D’Agostino Jr R, Howard G, et al. Chronic subclinical inflammation as part of the insulin resistance syndrome: The Insulin Resistance Atherosclerosis Study (IRAS). Circulation. 2000;102(1):42–7.PubMed
21.
Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with cardiovascular disease. Nature. 2006;444(7121):875–80.PubMed
22.
Persson M, Hedblad B, Nelson JJ, Berglund G. Elevated Lp-PLA2 levels add prognostic information to the metabolic syndrome on incidence of cardiovascular events among middle-aged nondiabetic subjects. Arterioscler Thromb Vasc Biol. 2007;27(6):1411–6.PubMed
23.
Han TS, Sattar N, Williams K, et al. Prospective study of C-reactive protein in relation to the development of diabetes and metabolic syndrome in the Mexico City Diabetes Study. Diabetes Care. 2002;25(11):2016–21.PubMed
24.
Laaksonen DE, Niskanen L, Nyyssonen K, et al. C-reactive protein and the development of the metabolic syndrome and diabetes in middle-aged men. Diabetologia. 2004;47(8):1403–10.PubMed
25.
Onat A, Ozhan H, Erbilen E, et al. Independent prediction of metabolic syndrome by plasma fibrinogen in men, and predictors of elevated levels. Int J Cardiol. 2009;135(2):211–7.PubMed
26.
Onat A, Can G, Hergenc G, et al. Serum apolipoprotein B predicts dyslipidemia, metabolic syndrome and, in women, hypertension and diabetes, independent of markers of central obesity and inflammation. Int J Obes (Lond). 2007;31(7):1119–25.
27.
Onat A, Uyarel H, Hergenc G, et al. Serum uric acid is a determinant of metabolic syndrome in a population-based study. Am J Hypertens. 2006;19(10):1055–62.PubMed
28.
Rubin D, Claas S, Pfeuffer M, et al. s-ICAM-1 and s-VCAM-1 in healthy men are strongly associated with traits of the metabolic syndrome, becoming evident in the postprandial response to a lipid-rich meal. Lipids Health Dis. 2008;732.
29.
Ruotolo G, Howard BV. Dyslipidemia of the metabolic syndrome. Curr Cardiol Rep. 2002;4(6):494–500.PubMed
30.
Onat A, Hergenc G. Low-grade inflammation, and dysfunction of high-density lipoprotein and its apolipoproteins as a major driver of cardiometabolic risk. Metabolism. 2011;60(4):499–512.PubMed
31.
Frigolet ME, Torres N, Tovar AR. The renin-angiotensin system in adipose tissue and its metabolic consequences during obesity. J Nutr Biochem. 2013;24(12):2003–15.PubMed
32.
Bassi N, Karagodin I, Wang S, et al. Lifestyle Modification for Metabolic Syndrome: A Systematic Review. Am J Med. 2014;
33.
Gerstein HC, Mohan V, Avezum A, et al. Long-term effect of rosiglitazone and/or ramipril on the incidence of diabetes. Diabetologia. 2011;54(3):487–95.PubMed
34.
Orchard TJ, Temprosa M, Goldberg R, et al. The effect of metformin and intensive lifestyle intervention on the metabolic syndrome: The Diabetes Prevention Program randomized trial. Ann Intern Med. 2005;142(8):611–9.PubMedCentralPubMed
35.
DeFronzo RA, Tripathy D, Schwenke DC, et al. Pioglitazone for diabetes prevention in impaired glucose tolerance. N Engl J Med. 2011;364(12):1104–15.PubMed
36.
Razavizade M, Jamali R, Arj A, et al. The effect of pioglitazone and metformin on liver function tests, insulin resistance, and liver fat content in nonalcoholic Fatty liver disease: a randomized double blinded clinical trial. Hepat Mon. 2013;13(5):e9270.PubMedCentralPubMed
37.
Colhoun HM, Betteridge DJ, Durrington PN, et al. Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the Collaborative Atorvastatin Diabetes Study (CARDS): Multicentre randomised placebo-controlled trial. Lancet. 2004;364(9435):685–96.PubMed
38.
Tenenbaum A, Fisman EZ. Fibrates are an essential part of modern anti-dyslipidemic arsenal: spotlight on atherogenic dyslipidemia and residual risk reduction. Cardiovasc Diabetol. 2012;11125.
39.
Vilsboll T, Christensen M, Junker AE, Knop FK, Gluud LL. Effects of glucagon-like peptide-1 receptor agonists on weight loss: systematic review and meta-analyses of randomised controlled trials. BMJ. 2012;344d7771.
40.
Bolinder J, Ljunggren O, Kullberg J, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J Clin Endocrinol Metab. 2012;97(3):1020–31.PubMed
41.
Chrysant SG, Chrysant GS, Chrysant C, Shiraz M. The treatment of cardiovascular disease continuum: focus on prevention and RAS blockade. Curr Clin Pharmacol. 2010;5(2):89–95.PubMed
42.
Thompson PD, Zimet R, Forbes WP, Zhang P. Meta-analysis of results from eight randomized, placebo-controlled trials on the effect of cilostazol on patients with intermittent claudication. Am J Cardiol. 2002;90(12):1314–9.PubMed
43.
Smith SR, Weissman NJ, Anderson CM, et al. Multicenter, placebo-controlled trial of lorcaserin for weight management. N Engl J Med. 2010;363(3):245–56.PubMed
44.
Coomans CP, Geerling JJ, van den Berg SA, et al. The insulin sensitizing effect of topiramate involves KATP channel activation in the central nervous system. Br J Pharmacol. 2013;170(4):908–18.PubMedCentralPubMed
45.
Apovian CM, Aronne L, Rubino D, et al. A randomized, phase 3 trial of naltrexone SR/bupropion SR on weight and obesity-related risk factors (COR-II). Obesity (Silver Spring). 2013;21(5):935–43.
46.
Lehrke M, Lazar MA. The many faces of PPARgamma. Cell. 2005;123(6):993–9.PubMed
47.
Zambon A, Gervois P, Pauletto P, Fruchart JC, Staels B. Modulation of hepatic inflammatory risk markers of cardiovascular diseases by PPAR-alpha activators: clinical and experimental evidence. Arterioscler Thromb Vasc Biol. 2006;26(5):977–86.PubMed
48.
Borghi C, Santi F. Fixed combination of lercanidipine and enalapril in the management of hypertension: focus on patient preference and adherence. Patient Prefer Adherence. 2012;6449–55
49.
Nauck MA. Incretin-based therapies for type 2 diabetes mellitus: properties, functions, and clinical implications. Am J Med. 2011;124(1 Suppl):S3–18.PubMed
50.
Gerich JE. Role of the kidney in normal glucose homeostasis and in the hyperglycaemia of diabetes mellitus: Therapeutic implications. Diabet Med. 2010;27(2):136–42.PubMed
51.
Hainer V, Hainerova IA. Do we need anti-obesity drugs? Diabetes Metab Res Rev. 2012;28(Suppl):28–20.
52.
Van Gaal LF, Rissanen AM, Scheen AJ, Ziegler O, Rossner S. Effects of the cannabinoid-1 receptor blocker rimonabant on weight reduction and cardiovascular risk factors in overweight patients: 1-year experience from the RIO-Europe study. Lancet. 2005;365(9468):1389–97.PubMed
53.
Despres JP, Golay A, Sjostrom L. Effects of rimonabant on metabolic risk factors in overweight patients with dyslipidemia. N Engl J Med. 2005;353(20):2121–34.PubMed
54.
Adan RA. Mechanisms underlying current and future anti-obesity drugs. Trends Neurosci. 2013;36(2):133–40.PubMed
55.
Brashier DB, Sharma AK, Dahiya N, Singh SK, Khadka A. Lorcaserin: A novel antiobesity drug. J Pharmacol Pharmacother. 2014;5(2):175–8.PubMedCentralPubMed
56.
Smith SM, Meyer M, Trinkley KE. Phentermine/topiramate for the treatment of obesity. Ann Pharmacother. 2013;47(3):340–9.PubMed
57.
Antel J, Hebebrand J. Weight-reducing side effects of the antiepileptic agents topiramate and zonisamide. Handb Exp Pharmacol. 2012;209:433–66.PubMed
58.
Hunt S, Russell A, Smithson WH, et al. Topiramate in pregnancy: Preliminary experience from the UK Epilepsy and Pregnancy Register. Neurology. 2008;71(4):272–6.PubMed
59.
Billes SK, Sinnayah P, Cowley MA. Naltrexone/bupropion for obesity: an investigational combination pharmacotherapy for weight loss. Pharmacol Res. 2014;841–11.
60.
Gupta D, Jetton TL, Mortensen RM, et al. In vivo and in vitro studies of a functional peroxisome proliferator-activated receptor gamma response element in the mouse pdx-1 promoter. J Biol Chem. 2008;283(47):32462–70.PubMedCentralPubMed
61.
Ackermann AM, Gannon M. Molecular regulation of pancreatic beta-cell mass development, maintenance, and expansion. J Mol Endocrinol. 2007;38(1–2):193–206.PubMed
62.
Stoffers DA, Thomas MK, Habener JF. Homeodomain protein IDX-1: A master regulator of pancreas development and insulin gene expression. Trends Endocrinol Metab. 1997;8(4):145–51.PubMed
63.
Gastaldelli A, Ferrannini E, Miyazaki Y, et al. Thiazolidinediones improve beta-cell function in type 2 diabetic patients. Am J Physiol Endocrinol Metab. 2007;292(3):E871–83.PubMed
64.
Matsui J, Terauchi Y, Kubota N, et al. Pioglitazone reduces islet triglyceride content and restores impaired glucose-stimulated insulin secretion in heterozygous peroxisome proliferator-activated receptor-gamma-deficient mice on a high-fat diet. Diabetes. 2004;53(11):2844–54.PubMed
65.
Lupi R, Del GS, Marselli L, et al. Rosiglitazone prevents the impairment of human islet function induced by fatty acids: evidence for a role of PPARgamma2 in the modulation of insulin secretion. Am J Physiol Endocrinol Metab. 2004;286(4):E560–7.PubMed
66.
Haataja L, Gurlo T, Huang CJ, Butler PC. Islet amyloid in type 2 diabetes, and the toxic oligomer hypothesis. Endocr Rev. 2008;29(3):303–16.PubMedCentralPubMed
67.
Kahn SE, Haffner SM, Heise MA, et al. Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl J Med. 2006;355(23):2427–43.PubMed
68.
Cornier MA, Dabelea D, Hernandez TL, et al. The metabolic syndrome. Endocr Rev. 2008;29(7):777–822.PubMed
69.
Kadowaki T, Yamauchi T, Kubota N, et al. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest. 2006;116(7):1784–92.PubMedCentralPubMed
70.
Samaha FF, Szapary PO, Iqbal N, et al. Effects of rosiglitazone on lipids, adipokines, and inflammatory markers in nondiabetic patients with low high-density lipoprotein cholesterol and metabolic syndrome. Arterioscler Thromb Vasc Biol. 2006;26(3):624–30.PubMed
71.
Singh S, Loke YK, Furberg CD. Long-term risk of cardiovascular events with rosiglitazone: a meta-analysis. JAMA. 2007;298(10):1189–95.PubMed
72.
Forman LM, Simmons DA, Diamond RH. Hepatic failure in a patient taking rosiglitazone. Ann Intern Med. 2000;132(2):118–21.PubMed
73.
Meier C, Kraenzlin ME, Bodmer M, et al. Use of thiazolidinediones and fracture risk. Arch Intern Med. 2008;168(8):820–5.PubMed
74.
Inzucchi SE, Bergenstal RM, Buse JB, et al. Management of hyperglycemia in type 2 diabetes: A patient-centered approach: Position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2012;35(6):1364–79.PubMedCentralPubMed
75.
Arunachalam G, Samuel SM, Marei I, Ding H, Triggle CR. Metformin modulates hyperglycaemia-induced endothelial senescence and apoptosis through SIRT1. Br J Pharmacol. 2014;171(2):523–35.PubMedCentralPubMed
76.
Krysiak R, Okopien B. Lymphocyte-suppressing and systemic anti-inflammatory effects of high-dose metformin in simvastatin-treated patients with impaired fasting glucose. Atherosclerosis. 2012;225(2):403–7.PubMed
77.
Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature. 1990;343(6257):425–30.PubMed
78.
Charlton-Menys V, Durrington PN. Human cholesterol metabolism and therapeutic molecules. Exp Physiol. 2008;93(1):27–42.PubMed
79.
Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005;366(9493):1267–78.PubMed
80.
Stalenhoef AF, Ballantyne CM, Sarti C, et al. A comparative study with rosuvastatin in subjects with metabolic syndrome: results of the COMETS study. Eur Heart J. 2005;26(24):2664–72.PubMed
81.
Stender S, Schuster H, Barter P, Watkins C, Kallend D. Comparison of rosuvastatin with atorvastatin, simvastatin and pravastatin in achieving cholesterol goals and improving plasma lipids in hypercholesterolaemic patients with or without the metabolic syndrome in the MERCURY I trial. Diabetes Obes Metab. 2005;7(4):430–8.PubMed
82.
Deedwania PC, Hunninghake DB, Bays HE, et al. Effects of rosuvastatin, atorvastatin, simvastatin, and pravastatin on atherogenic dyslipidemia in patients with characteristics of the metabolic syndrome. Am J Cardiol. 2005;95(3):360–6.PubMed
83.
Mihaylova B, Emberson J, Blackwell L, et al. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet. 2012;380(9841):581–90.PubMed
84.
Koo BK. Statin for the primary prevention of cardiovascular disease in patients with diabetes mellitus. Diabetes Metab J. 2014;38(1):32–4.PubMedCentralPubMed
85.
LaRosa JC, Grundy SM, Waters DD, et al. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med. 2005;352(14):1425–35.PubMed
86.
Pyorala K, Ballantyne CM, Gumbiner B, et al. Reduction of cardiovascular events by simvastatin in nondiabetic coronary heart disease patients with and without the metabolic syndrome: Subgroup analyses of the Scandinavian Simvastatin Survival Study (4S). Diabetes Care. 2004;27(7):1735–40.PubMed
87.
Liao JK. Beyond lipid lowering: The role of statins in vascular protection. Int J Cardiol. 2002;86(1):5–18.PubMed
88.
Meyer-Sabellek W, Brasch H. Atherosclerosis, inflammation, leukocyte function and the effect of statins. J Hypertens. 2006;24(12):2349–51.PubMed
89.
Goff Jr DC, Lloyd-Jones DM, Bennett G, et al. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation. 2014;129(25 Suppl 2):S49–73.PubMed
90.
Preiss D, Seshasai SR, Welsh P, et al. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: A meta-analysis. JAMA. 2011;305(24):2556–64.PubMed
91.
Sattar N, Preiss D, Murray HM, et al. Statins and risk of incident diabetes: A collaborative meta-analysis of randomised statin trials. Lancet. 2010;375(9716):735–42.PubMed
92.
Chamberlain LH. Inhibition of isoprenoid biosynthesis causes insulin resistance in 3 T3-L1 adipocytes. FEBS Lett. 2001;507(3):357–61.PubMed
93.
Kanda M, Satoh K, Ichihara K. Effects of atorvastatin and pravastatin on glucose tolerance in diabetic rats mildly induced by streptozotocin. Biol Pharm Bull. 2003;26(12):1681–4.PubMed
94.
Ridker PM, Pradhan A, MacFadyen JG, Libby P, Glynn RJ. Cardiovascular benefits and diabetes risks of statin therapy in primary prevention: An analysis from the JUPITER trial. Lancet. 2012;380(9841):565–71.PubMedCentralPubMed
95.
Sugiyama T, Tsugawa Y, Tseng CH, Kobayashi Y, Shapiro MF. Different time trends of caloric and fat intake between statin users and nonusers among US adults: Gluttony in the time of statins? JAMA Intern Med. 2014;174(7):1038–45.PubMed
96.
Lim S, Sakuma I, Quon MJ, Koh KK. Potentially important considerations in choosing specific statin treatments to reduce overall morbidity and mortality. Int J Cardiol. 2013;167(5):1696–702.PubMed
97.
Keech A, Simes RJ, Barter P, et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): Randomised controlled trial. Lancet. 2005;366(9500):1849–61.PubMed
98.
Guilar-Salinas CA, Fanghanel-Salmon G, Meza E, et al. Ciprofibrate versus gemfibrozil in the treatment of mixed hyperlipidemias: An open-label, multicenter study. Metabolism. 2001;50(6):729–33.
99.
Klosiewicz-Latoszek L, Szostak WB. Comparative studies on the influence of different fibrates on serum lipoproteins in endogenous hyperlipoproteinaemia. Eur J Clin Pharmacol. 1991;40(1):33–41.PubMed
100.
Packard KA, Backes JM, Lenz TL, et al. Comparison of gemfibrozil and fenofibrate in patients with dyslipidemic coronary heart disease. Pharmacotherapy. 2002;22(12):1527–32.PubMed
101.
Kilicarslan A, Yavuz B, Guven GS, et al. Fenofibrate improves endothelial function and decreases thrombin-activatable fibrinolysis inhibitor concentration in metabolic syndrome. Blood Coagul Fibrinolysis. 2008;19(4):310–4.PubMed
102.
Lim S, Park YM, Sakuma I, Koh KK. How to control residual cardiovascular risk despite statin treatment: Focusing on HDL-cholesterol. Int J Cardiol. 2013;166(1):8–14.PubMed
103.
Watanabe S, Tagawa T, Yamakawa K, Shimabukuro M, Ueda S. Inhibition of the renin-angiotensin system prevents free fatty acid-induced acute endothelial dysfunction in humans. Arterioscler Thromb Vasc Biol. 2005;25(11):2376–80.PubMed
104.
Henriksen EJ, Prasannarong M. The role of the renin-angiotensin system in the development of insulin resistance in skeletal muscle. Mol Cell Endocrinol. 2013;378(1–2):15–22.PubMed
105.
Shatanawi A, Romero MJ, Iddings JA, et al. Angiotensin II-induced vascular endothelial dysfunction through RhoA/Rho kinase/p38 mitogen-activated protein kinase/arginase pathway. Am J Physiol Cell Physiol. 2011;300(5):C1181–92.PubMedCentralPubMed
106.
Yusuf S, Teo KK, Pogue J, et al. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008;358(15):1547–59.PubMed
107.
Patel A, MacMahon S, Chalmers J, et al. Effects of a fixed combination of perindopril and indapamide on macrovascular and microvascular outcomes in patients with type 2 diabetes mellitus (the ADVANCE trial): A randomised controlled trial. Lancet. 2007;370(9590):829–40.PubMed
108.
Contreras F, de la Parte MA, Cabrera J, et al. Role of angiotensin II AT1 receptor blockers in the treatment of arterial hypertension. Am J Ther. 2003;10(6):401–8.PubMed
109.
Wan Y, Kurosaki T, Huang XY. Tyrosine kinases in activation of the MAP kinase cascade by G-protein-coupled receptors. Nature. 1996;380(6574):541–4.PubMed
110.
Yusuf S, Sleight P, Pogue J, et al. 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(3):145–53.PubMed
111.
Geng DF, Jin DM, Wu W, Liang YD, Wang JF. Angiotensin converting enzyme inhibitors for prevention of new-onset type 2 diabetes mellitus: A meta-analysis of 72,128 patients. Int J Cardiol. 2013;167(6):2605–10.PubMed
112.
Reid JL. Molecular-specific effects of angiotensin II antagonists: Clinical relevance to treating hypertension? J Renin-Angiotensin-Aldosterone Syst. 2005;6(1):15–24.PubMed
113.
Kohlstedt K, Gershome C, Trouvain C, et al. Angiotensin-converting enzyme (ACE) inhibitors modulate cellular retinol-binding protein 1 and adiponectin expression in adipocytes via the ACE-dependent signaling cascade. Mol Pharmacol. 2009;75(3):685–92.PubMed
114.
Chin BS, Langford NJ, Nuttall SL, et al. Anti-oxidative properties of beta-blockers and angiotensin-converting enzyme inhibitors in congestive heart failure. Eur J Heart Fail. 2003;5(2):171–4.PubMed
115.
Manabe S, Okura T, Watanabe S, Fukuoka T, Higaki J. Effects of angiotensin II receptor blockade with valsartan on pro-inflammatory cytokines in patients with essential hypertension. J Cardiovasc Pharmacol. 2005;46(6):735–9.PubMed
116.
Edwards KM, Ziegler MG, Mills PJ. The potential anti-inflammatory benefits of improving physical fitness in hypertension. J Hypertens. 2007;25(8):1533–42.PubMed
117.
Borghi C, Cicero AF. The role of irbesartan in the treatment of patients with hypertension: A comprehensive and practical review. High Blood Press Cardiovasc Prev. 2012;19(1):19–31.PubMed
118.
Zreikat HH, Harpe SE, Slattum PW, et al. Effect of Renin-Angiotensin system inhibition on cardiovascular events in older hypertensive patients with metabolic syndrome. Metabolism. 2014;63(3):392–9.PubMed
119.
Gutzwiller JP, Drewe J, Goke B, et al. Glucagon-like peptide-1 promotes satiety and reduces food intake in patients with diabetes mellitus type 2. Am J Physiol. 1999;276(5 Pt 2):R1541–4.PubMed
120.
Turton MD, O’Shea D, Gunn I, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379(6560):69–72.PubMed
121.
Meier JJ. GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat Rev Endocrinol. 2012;8(12):728–42.PubMed
122.
Buteau J. GLP-1 receptor signaling: effects on pancreatic beta-cell proliferation and survival. Diabetes Metab. 2008;34 Suppl 2:S73–7.PubMed
123.
Holz GG, Leech CA, Habener JF. Activation of a cAMP-regulated Ca(2+)-signaling pathway in pancreatic beta-cells by the insulinotropic hormone glucagon-like peptide-1. J Biol Chem. 1995;270(30):17749–57.PubMedCentralPubMed
124.
Yusta B, Baggio LL, Estall JL, et al. GLP-1 receptor activation improves beta cell function and survival following induction of endoplasmic reticulum stress. Cell Metab. 2006;4(5):391–406.PubMed
125.
Astrup A, Rossner S, Van GL, et al. Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet. 2009;374(9701):1606–16.PubMed
126.
DeFronzo RA, Davidson JA, Del PS. The role of the kidneys in glucose homeostasis: a new path towards normalizing glycaemia. Diabetes Obes Metab. 2012;14(1):5–14.PubMed
127.
List JF, Woo V, Morales E, Tang W, Fiedorek FT. Sodium-glucose cotransport inhibition with dapagliflozin in type 2 diabetes. Diabetes Care. 2009;32(4):650–7.PubMedCentralPubMed
128.
Strojek K, Yoon KH, Hruba V, et al. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with glimepiride: A randomized, 24-week, double-blind, placebo-controlled trial. Diabetes Obes Metab. 2011;13(10):928–38.PubMed
129.
Ferrannini E, Solini A. SGLT2 inhibition in diabetes mellitus: rationale and clinical prospects. Nat Rev Endocrinol. 2012;8(8):495–502.PubMed
130.
Ferrannini E, Ramos SJ, Salsali A, Tang W, List JF. Dapagliflozin monotherapy in type 2 diabetic patients with inadequate glycemic control by diet and exercise: A randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care. 2010;33(10):2217–24.PubMedCentralPubMed
131.
Henry RR, Murray AV, Marmolejo MH, et al. Dapagliflozin, metformin XR, or both: Initial pharmacotherapy for type 2 diabetes, a randomised controlled trial. Int J Clin Pract. 2012;66(5):446–56.PubMed
132.
Jurczak MJ, Lee HY, Birkenfeld AL, et al. SGLT2 deletion improves glucose homeostasis and preserves pancreatic beta-cell function. Diabetes. 2011;60(3):890–8.PubMedCentralPubMed
133.
Bailey CJ, Gross JL, Pieters A, Bastien A, List JF. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with metformin: a randomised, double-blind, placebo-controlled trial. Lancet. 2010;375(9733):2223–33.PubMed
134.
Nauck MA, Del PS, Meier JJ, et al. Dapagliflozin versus glipizide as add-on therapy in patients with type 2 diabetes who have inadequate glycemic control with metformin: A randomized, 52-week, double-blind, active-controlled noninferiority trial. Diabetes Care. 2011;34(9):2015–22.PubMedCentralPubMed
135.
Rosenstock J, Aggarwal N, Polidori D, et al. Dose-ranging effects of canagliflozin, a sodium-glucose cotransporter 2 inhibitor, as add-on to metformin in subjects with type 2 diabetes. Diabetes Care. 2012;35(6):1232–8.PubMedCentralPubMed
136.
Jung CH, Jang JE, Park JY. A novel therapeutic agent for type 2 diabetes mellitus: SGLT2 inhibitor. Diabetes Metab J. 2014;38(4):261–73.PubMedCentralPubMed
137.
Suzuki K, Uchida K, Nakanishi N, Hattori Y. Cilostazol activates AMP-activated protein kinase and restores endothelial function in diabetes. Am J Hypertens. 2008;21(4):451–7.PubMed
138.
Aoki C, Hattori Y, Tomizawa A, Jojima T, Kasai K. Anti-inflammatory role of cilostazol in vascular smooth muscle cells in vitro and in vivo. J Atheroscler Thromb. 2010;17(5):503–9.PubMed
139.
Fujita K, Nozaki Y, Wada K, et al. Effectiveness of antiplatelet drugs against experimental non-alcoholic fatty liver disease. Gut. 2008;57(11):1583–91.PubMed
140.
Tani T, Uehara K, Sudo T, et al. Cilostazol, a selective type III phosphodiesterase inhibitor, decreases triglyceride and increases HDL cholesterol levels by increasing lipoprotein lipase activity in rats. Atherosclerosis. 2000;152(2):299–305.PubMed
141.
Alexopoulos N, Katritsis D, Raggi P. Visceral adipose tissue as a source of inflammation and promoter of atherosclerosis. Atherosclerosis. 2014;233(1):104–12.PubMed
142.
Gustafson B, Hammarstedt A, Andersson CX, Smith U. Inflamed adipose tissue: A culprit underlying the metabolic syndrome and atherosclerosis. Arterioscler Thromb Vasc Biol. 2007;27(11):2276–83.PubMed

Be confident that your patient care is up to date

Medicine Matters is being incorporated into Springer Medicine, our new medical education platform. 

Alongside the news coverage and expert commentary you have come to expect from Medicine Matters diabetes, Springer Medicine's complimentary membership also provides access to articles from renowned journals and a broad range of Continuing Medical Education programs. Create your free account »