You are on page 1of 7

Journal of the American College of Cardiology 2005 by the American College of Cardiology Foundation Published by Elsevier Inc.

Vol. 45, No. 12, 2005 ISSN 0735-1097/05/$30.00 doi:10.1016/j.jacc.2005.03.041

CLINICAL RESEARCH

Clinical Trial

Improvement of Cardiovascular Risk Markers by Pioglitazone Is Independent From Glycemic Control


Results From the Pioneer Study
Andreas Pftzner, MD, PHD,* Nikolaus Marx, MD, Georg Lbben, MD, Matthias Langenfeld, MD,* Daniel Walcher, MD, Thomas Konrad, MD, Thomas Forst, MD* Mainz, Rheinbach, Ulm, Aachen, and Frankfurt, Germany
This study was performed to assess whether the anti-inammatory and antiatherogenic effects of pioglitazone suggested by animal experiments are reproducible in man and independent from improvements in metabolic control. BACKGROUND Type 2 diabetes is associated with increased cardiovascular risk. METHODS A total of 192 patients were enrolled into a six-month, prospective, open-label, controlled clinical study. They were randomized to receive either pioglitazone (45 mg) or glimepiride (1 to 6 mg, with the intent to optimize therapy). Biochemical and clinical markers to assess therapeutic effects included HbA1c, fasting glucose, insulin, adiponectin, lipids, highsensitivity C-reactive protein (hsCRP), intracellular adhesion molecule, vascular cell adhesion molecule, vascular endothelial growth factor, brinogen, von Willebrand factor, matrix metalloproteinase (MMP)-9, monocyte chemoattractant protein (MCP)-1, soluble CD40 ligand, and carotid intima-media thickness (IMT). RESULTS The study was completed by 173 patients (66 female, 107 male; age [ SD]: 63 8 years; disease duration: 7.2 7.2 years; HbA1c: 7.5 0.9%; pioglitazone arm: 89 patients). A comparable reduction in HbA1c was seen in both groups (p 0.001). In the pioglitazone group, reductions were observed for glucose (p 0.001 vs. glimepiride group at end point), insulin (p 0.001), low-density lipoprotein/high-density lipoprotein ratio (p 0.001), hsCRP (p 0.05), MMP-9 (p 0.05), MCP-1 (p 0.05), and carotid IMT (p 0.001), and an increase was seen in high-density lipoprotein (p 0.001) and adiponectin (p 0.001). Spearman ranks analysis revealed only one correlation between the reduction in cardiovascular risk parameters and the improvement in the metabolic parameters (MMP-9 and fasting blood glucose, p 0.05) CONCLUSIONS This prospective study gives evidence of an anti-inammatory and antiatherogenic effect of pioglitazone versus glimepiride. This effect is independent from blood glucose control and may be attributed to peroxisome proliferator-activated receptor gamma activation. (J Am Coll Cardiol 2005;45:192531) 2005 by the American College of Cardiology Foundation OBJECTIVES

Patients with type 2 diabetes mellitus exhibit an increased propensity to develop extensive arteriosclerosis with its sequelae, unstable angina pectoris and acute myocardial infarction (1,2). Over the last years, experimental data have illuminated the role of inammation in atherogenesis, while clinical studies have shown that this concept of inammation in arteriosclerosis applies directly to human patients (3). As such, increased serum levels of inammatory biomarkers of arteriosclerosis, like C-reactive protein, cytokines, like tumor necrosis factor-alpha or interleukin-6, as well as novel markers like monocyte chemoattractant protein (MCP)-1, soluble CD40 ligand (sCD40L), and matrix metalloproFrom the *IKFEInstitute for Clinical Research and Development, Mainz, Germany; University of Applied Sciences, Rheinbach, Germany; University Hospital, Ulm, Germany; Takeda Pharma GmbH, Aachen, Germany; and ISF Institute for Metabolic Research, Frankfurt, Germany. The study has been sponsored by Takeda Pharma, Germany. Dr. Pftzner received research grants from Takeda. Dr. Forst received research grants and speaker fees from Takeda. Dr. Konrad received research grants and speaker fees from Takeda. Dr. Marx received research grants and speaker fees from Takeda. Dr. Lbben is employed by Takeda Pharma. Manuscript received October 12, 2004; revised manuscript received February 14, 2005, accepted March 10, 2005.

teinases (MMP) have been shown to predict cardiovascular risk and seem to reect the overall burden of vascular disease in patients. Interestingly, some of these markers are elevated in patients with type 2 diabetes and insulin resistance, indicating a pivotal role of inammation in this metabolic disorder (4 6). Recent data suggest that the release of inammatory mediators like tumor necrosis factor-alpha and interleukin-6 from the visceral adipose tissue as well as an activation of vascular cells itself contribute to the inammatory state in these patients with metabolic syndrome. Therefore, enhanced serum levels of sCD40L may reect endothelial and platelet activation in diabetic subjects, while increased MMP-9 levels suggest the presence of unstable plaques with activated monocytes/macrophages (711). Moreover, elevated soluble adhesion molecules like soluble intracellular adhesion molecule (sICAM) and soluble vascular cell adhesion molecule (sVCAM) are markers of endothelial dysfunction in these patients. Given the increased risk of diabetic patients for macrovascular events, therapeutic strategies that limit inammation in the vessel wall and reduce serum levels of inammatory surrogate

1926

Pftzner et al. Cardiovascular Risk Improvement by Pioglitazone

JACC Vol. 45, No. 12, 2005 June 21, 2005:192531

Abbreviations and Acronyms HDL high-density lipoprotein hsCRP high-sensitivity C-reactive protein ICAM intracellular adhesion molecule IMT intima-media thickness LDL low-density lipoprotein MCP monocyte chemoattractant protein MMP matrix metalloproteinase PPAR peroxisome proliferator-activated receptor gamma PROactive Prospective Pioglitazone Clinical Trial in Macrovascular Events sCD40L soluble CD40 ligand TZD thiazolidinedione VCAM vascular cell adhesion molecule VEGF vascular endothelial growth factor

parameters have been considered a promising tool to inuence vascular disease in this high-risk population (12,13). Recent experimental and clinical data suggest that a novel group of antidiabetic agents, thiazolidinediones (TZDs), like pioglitazone and rosiglitazone, may exhibit antiinammatory properties in the vessel wall (14,15). These agents act via the nuclear transcription factor peroxisome proliferator-activated receptor gamma (PPAR ) and, in addition to their metabolic action, have been shown to regulate the expression of various target genes in vascular cells in vitro and in vivo, subsequently limiting inammatory cell activation and lesion formation during atherogenesis. Furthermore, clinical data suggest that TZDs reduce inammatory biomarkers of arteriosclerosis, like C-reactive protein or sCD40L, in treated patients, thus potentially modulating their cardiovascular risk (16 19). Still, most of these studies were placebo-controlled and, as such, did not allow the dissection of metabolic from nonmetabolic TZDs effects, because TZD treatment, compared to placebo, signicantly improved glucose metabolism in all of these studies. Therefore, we performed a six-month prospective, randomized, controlled trial to compare the effect of pioglitazone and sulfonylurea treatment on inammatory biomarkers of arteriosclerosis, attempting to achieve comparable improvement in blood glucose control in both treatment groups.

METHODS
The prospective randomized monocentric study was approved by the responsible ethics committee and was performed in accordance with the Declaration of Helsinki and the guidelines for good clinical practice. All patient examinations were performed at the Clinical Department of the Institute for Clinical Research and Development (IKFE), Mainz, Germany. After written informed consent was obtained, 192 orally treated patients with type 2 diabetes without prior TZD treatment were enrolled into the trial. After randomization, they either received a xed dose of pioglitazone (45 mg/day) in the morning or glimepiride (1

to 6 mg/day), titrated for optimal glycemic control. Inclusion criteria included an age of 40 to 75 years, HbA1c: 6.6% to 9.9%, absence of signicant hepatic or renal disease, absence of congestive heart failure (New York Heart Association functional class II to IV), no cigarette smoking, and no known carotid artery disease. All study measurements were obtained at study entry and after 26 2 weeks. In order to improve metabolic control, individual medical advice was given to every patient throughout the study. In the pioglitazone group, other additional oral antidiabetic therapy was permitted except for metformin, while only TZDs were excluded for additional treatment in the glimepiride group. All blood draws and measurements were performed in the morning after fasting of the patients from midnight onward. Biochemical parameters. HbA1c was determined by means of high-pressure liquid chromatography (Adams TMA1c, Menarini Diagnostics, Florence, Italy). Therapy response was dened as an absolute decrease in HbA1c from baseline by at least 0.6% (normal reference range: 4.2% to 6.0%). Glucose was assessed using a standard glucose oxidase reference method (Ruhrtal Labortechnik, Mhnesee-Delecke, Germany), and lipids (total cholesterol, low-density lipoprotein [LDL], high-density lipoprotein [HDL], triglycerides) were measured by means of standard dry chemistry methods (OSR, Olympus Diagnostica, Hamburg, Germany). Insulin was measured using a chemiluminescence method (Sciema, Mainz, Germany), PAI-I by ELISA (American Diagnostica, Pfungstadt, Germany), endothelin by ELISA (Biomedica, Vienna, Austria), and adiponectin by radioimmunoassay (Linco, St. Charles, Missouri); ELISAs from R&D Systems (Wiesbaden, Germany) were used for the determination of the following parameters: ICAM, VCAM, vascular endothelial growth factor [VEGF], MMP-9, MCP-1. The sCD40L determinations were also performed by ELISA (Bender Medsystems, Vienna, Austria). Turbimetric methods were used for the following parameters: high-sensitivity C-reactive protein (hsCRP) (Olympus, Hamburg, Germany), brinogen (Dade Behring, Schwalbach, Germany), and vonWillebrand factor (Instrumentation Laboratory GmbH, Kirchheim, Germany). High-sensitive C-reactive protein changes 15 mg/l during the observation period were attributed to other inammatory processes (u, cold, and so on) and were eliminated before analysis. Carotid intima-media thickness (IMT). Carotid IMT was evaluated at all time points by a single operator with high-resolution B-mode ultrasound on a single machine (Caris Plus, Esaote SpA, Genoa, Italy) with a 10-MHz linear array transducer (LA 523). All recordings were performed in a standardized way, and readings were analyzed by a physician blinded to patient prole and treatment assignment as described previously (20,21). Statistical analysis. The analysis of efcacy is based on the intention-to-treat population, which consists of all patients

JACC Vol. 45, No. 12, 2005 June 21, 2005:192531

Pftzner et al. Cardiovascular Risk Improvement by Pioglitazone

1927

who were treated and provided assessment of the laboratory parameters at baseline and at end point of the study. All analyses were performed in an exploratory sense with appropriate parametrical and nonparametrical methods. Treatment groups were compared at baseline by using a Wilcoxon rank sum test for continuous variables and chisquare for categorical variables. Changes from baseline were evaluated by using analysis of covariance (ANCOVA) models with treatment groups as factor and baseline values as covariate. The difference between treatment groups was assessed by using t test statistics for the hypothesis that treatment group is a relevant factor in the model. Spearman rank correlation coefcients were calculated to test for the independence of the obtained results. All p values 0.05 were interpreted as statistically signicant.

RESULTS
Of 192 patients, 179 were treated, and 173 were included into the intention-to-treat population. The most frequent reason for early termination was patient decision (seven in the pioglitazone group, three in the glimepiride group, p 0.226). The patient characteristics of the nal analysis group are given in Table 1. There were no signicant differences between the treatment groups with regard to demographic or treatment parameters. In total, 162 patients completed the protocol. Glimepiride was administered at an average daily dose of 2.7 1.6 mg. Both treatments were well tolerated, and no episode of severe hypoglycemia was recorded. Cardiac failure requiring hospitalization was reported for two patients in the pioglitazone group. Effects of pioglitazone and glimepiride on metabolic parameters. There were no differences in all metabolic parameters at baseline. In both groups an equal and significant improvement of HbA1c was observed (pioglitazone: 0.8 0.9%; glimepiride: 0.6 0.8%; p 0.001 vs. baseline in both groups, no signicant difference between the groups at baseline and end point). Fasting glucose decreased at a higher extent in the pioglitazone group as compared to the glimepiride group (pioglitazone: 17.8
Table 1. Patient Characteristics

33.5 mg/dl; glimepiride: 4.8 30.8 mg/dl, p 0.01). Fasting serum insulin concentrations decreased signicantly in the pioglitazone group ( 4.7 6.2 U/ml, p 0.001), while they remained unchanged in the glimepiride treatment arm (0.4 5.7 U/ml, p NS). A signicant increase of HDL cholesterol was seen in the pioglitazone group only (from 46 11 mg/dl to 54 13 mg/dl, p 0.001), while a decrease in total cholesterol was observed in the glimepiride group only (from 228 38 mg/dl to 215 39 mg/dl, p 0.001). Free fatty acids, triglycerides, and adiponectin improved signicantly in the pioglitazone group only. A summary of the effects of both treatment moieties on the metabolic markers is given in Table 2. Effects of pioglitazone and glimepiride on cardiovascular risk parameters. There were no differences in all cardiovascular risk parameters at baseline. For the remaining group, a signicant reduction of hsCRP was seen after six months of therapy in the pioglitazone arm ( 0.10 2.25 mg/dl, 29%, end point vs. baseline, p 0.001), while no such change could be seen during glimpiride treatment ( 0.24 4.04 mg/dl, 4%, p NS). Treatment with pioglitazone signicantly decreased MMP-9 and MCP-1 concentrations, while glimepiride did not induce changes in these parameters. At end point, there were signicant differences between the groups for both MCP-1 and MMP-9 in favor of pioglitazone (p 0.05 in both cases). No signicant changes or differences were observed for sCD40L, VEGF, ICAM, VCAM, brinogen, vonWillebrand factor, PAI-I, or endothelin-I. A summary of the values at baseline and end point for both treatment groups is given in Table 3, and the percent values for the signicantly changed parameters are given in Figure 1. While both groups were comparable at baseline, substantial regression of carotid IMT was seen in the pioglitazone treatment arm only ( 54 59 m, p 0.001 vs. baseline). The minor changes observed in the glimepiride group did not reach the level of statistical signicance ( 11 58 m, p 0.001 between the groups at end point). Spearman correlation revealed a correlation between MCP-1 and

Pioglitazone Group (n 89) Age (yrs) Gender (M:F) Ethnicity (Caucasian:other) Duration of diabetes (yrs) HbA1c (%) HOMA-IR (mmol U/ml/22.5) Body mass index (kg/m2) Systolic blood pressure (mm Hg) Statin treatment (n, %) ACE inhibitor/AT-1 antagonist (n, %) Antiplatelet therapy (n, %) Intima-media thickness (mm) Plaques (n, %)
ACE

Glimepiride Group (n 84) 63.0 7.4 52:32 81:3 6.9 6.5 7.44 0.89 5.8 3.7 31.8 4.3 148 20 13 (15%) 41 (49%) 26 (31%) 0.924 0.150 36 (43%)

p Value NS NS NS NS NS NS NS NS NS NS NS NS NS

62.2 8.4 55:34 88:1 7.4 7.9 7.52 0.85 6.2 4.1 31.7 5.0 149 21 18 (20%) 52 (58%) 25 (28%) 0.949 0.149 47 (53%)

angiotensin-converting enzyme; HOMA-IR Homeostatsis Model Assessment for insulin resistance.

1928

Pftzner et al. Cardiovascular Risk Improvement by Pioglitazone

JACC Vol. 45, No. 12, 2005 June 21, 2005:192531

Table 2. Changes in the Metabolic Parameters


Baseline Parameter HbA1c (%) Fasting serum glucose (mg/dl) Fasting serum insulin ( U/ml) HOMA-IR (mmol/l U/ml) BMI (kg/m2) Total cholesterol (mg/dl) LDL cholesterol (mg/dl) HDL cholesterol (mg/dl) LDL/HDL ratio Triglycerides (mg/dl) Free fatty acids (mmol/l) Adiponectin (mg/l)
*p

End Point (26 Weeks) Glimeperide (n 84) 7.44 144 16 5.8 31.8 228 137 46 3.2 202 0.55 6.5 0.89 34 9 3.7 4.3 38 25 14 0.9 111 0.18 3.4 Pioglitazone (n 89) 6.71 128 12 3.9 33.1 228 133 54 2.6 168 0.50 17.7 0.89 29 5 1.9 5.1 43 31 13 0.9* 102 0.20* 8.8 Glimepiride (n 84) 6.83 138 17 6.0 31.8 215 129 47 2.9 185 0.56 6.0 0.85 32 10 4.7 4.1 39 27 12 0.7 106 0.19 2.6

Pioglitazone (n 89) 7.52 147 17 6.2 31.7 227 136 46 3.1 190 0.54 6.7 0.85 37 9 4.1 5.0 41 29 11 0.9 109 0.21 3.4

p Value (Between Groups at End Point) p p p p p p p p p NS 0.01 0.001 0.001 0.001 0.005 NS 0.001 0.05 NS 0.05 0.001

0.05; p 0.005 vs. baseline; p 0.001 vs. baseline. BMI body mass index; HDL high-density lipoprotein; HOMA-IR

Homeostatsis Model Assessment for insulin resistance; LDL

low-density lipoprotein.

carotid IMT in the pioglitazone arm (p 0.05), while no correlation was seen between IMT reduction and hsCRP, MMP-9, or any of the other parameters. The observed changes were independent from the improvement of longterm glucose control, which is consistent with metabolic ndings by other groups (22). A stratication into therapy responders (reduction in HbA1c 0.6%) and nonresponders (reduction in HbA1c 0.6%) revealed no difference in the overall results (Fig. 2). Spearman correlation analyses between cardiovascular risk parameters and glycemic control parameters resulted in only one correlation (reduction in MMP-9 and reduction in fasting glucose control, p 0.05) among the multiple possible combinations (Table 4).

DISCUSSION
This prospective randomized controlled trial demonstrates signicant improvements of multiple cardiovascular risk markers during treatment with pioglitazone in comparison to glimepiride administration over six months. Because
Table 3. Changes in the Cardiovascular Risk Parameters
Baseline Parameter IMT (mm) hsCRP (mg/l) MCP-1 MMP-9 sCD40L ICAM (ng/ml) VCAM (ng/ml) Fibrinogen (g/l) VEGF (pg/ml) von Willebrand factor PAI-1 (ng/ml) Endothelin-I (pmol/l)
*p

metabolic control, as indicated by HbA1c values, was comparably improved in both treatment arms, the observed benecial effects of pioglitazone on cardiovascular risk markers are suggested to be independent from overall metabolic improvement but may rather be direct effects of PPAR activation. This hypothesis is supported by the fact that the observed changes were equally seen in therapy responders and nonresponders. In comparison to glimepiride, pioglitazone treatment led to a signicant increase in HDL cholesterol and adiponectin, decrease in fasting glucose and insulin, signicant higher reduction of triglycerides, and a signicant higher reduction in the LDL/HDL ratio. These pronounced differences between pioglitazone and sulfonylurea treatment have been consistently described by several groups in the literature (2326). As one explanation for these ndings, glyceroneogenesis has been recently identied as a target of TZDs in cultured adipocytes and fat tissues of Wistar rats. The activation of glyceroneogenesis by TZDs occurs mainly in visceral fat, the same fat depot that is specically

End Point (26 Weeks) Glimeperide (n 84) 0.924 3.26 388 354 2.61 310 820 3.02 409 106 46.3 0.50 0.150 4.31 116 179 2.48 78 446 0.93 235 41 7.4 1.69 Pioglitazone (n 89) 0.893 2.50 368 320 3.15 296 813 3.21 409 110 45.1 0.58 0.144 2.78 99* 122* 2.71 61 467 0.67 243 36 8.5 1.83 Glimepiride (n 84) 0.911 3.13 386 362 2.98 301 800 3.01 402 113 46.8 0.46 0.158 3.50 125 179 2.60 93 463 0.67 206 35 5.6 1.34

Pioglitazone (n 89) 0.949 3.49 397 373 2.89 299 809 3.23 415 110 45.3 0.60 0.149 3.26 111 147 2.21 60 374 1.04 245 41 7.9 1.67

p Value (Absolute Changes, Between Groups) p p p p 0.001 0.05 0.05 0.01 NS NS NS NS NS NS NS NS

0.005 vs. baseline; p 0.001 vs. baseline. hsCRP high-sensitivity C-reactive protein; ICAM intracellular adhesion molecule; IMT intima-media thickness; MCP-1 monocyte chemoattractant protein 1; MMP-9 matrix metalloproteinase 9; sCD40L soluble CD40 ligand; VCAM vascular cell adhesion molecule; VEGF vascular endothelial growth factor.

JACC Vol. 45, No. 12, 2005 June 21, 2005:192531

Pftzner et al. Cardiovascular Risk Improvement by Pioglitazone

1929

Table 4. Spearman Analysis of the Relations Between Metabolic and Macrovascular Risk Markers
HbA1c hsCRP MMP-9 MCP-1 0.4477 0.6238 0.6075 Fasting Glucose 0.6271 0.0379 0.2447 Fasting Insulin 0.0745 0.1190 0.9249 HOMA-IR 0.2178 0.0589 0.6829

Abbreviations as in Tables 2 and 3.

Figure 1. Mean of individual percent changes from baseline in selected biochemical and clinical cardiovascular risk markers. hsCRP highsensitivity C-reactive protein; IMT carotid intima media thickness; MCP monocyte chemoattractant protein; MMP matrix metalloproteinase.

implicated in the progression of obesity to type 2 diabetes. The main role of this metabolic pathway is to allow the re-esterication of fatty acids via a futile cycle in adipocytes, thus lowering fatty acid release into the plasma (27). Inammation plays an important role in arteriosclerosis, and measurement of hsCRP has become a novel but emerging tool for detecting individuals at high risk for plaque rupture. A randomized placebo-controlled study with different doses of rosiglitazone resulted in a decrease of hsCRP by 26.8% (4 mg) and 21.8% (8 mg) as compared to the placebo group after 26 weeks in a Caucasian study population (28). In another recent study performed in Japanese patients, treatment with 45 mg of pioglitazone signicantly reduced hsCRP by about 27% after three months of treatment, while no change occurred in the placebo comparator group. Independence from glucose metabolism was suggested by ANCOVA analysis (29). In our study, baseline values were much higher than in the

Figure 2. Stratication of percent changes in the cardiovascular risk markers according to therapeutic response (responders: absolute change in HbA1c 0.6%; nonresponders: absolute change in HbA1c 0.6%). Abbreviations as in Figure 1.

Japanese study. The decrease in hsCRP by 29% during pioglitazone treatment for the rst time compares directly to an equally effective antidiabetic comparator treatment that did not induce any hsCRP change. Substantial evidence supports a causal role for MCP-1 and its receptor, CCR2, in the recruitment of monocytes from the circulation into atherosclerotic lesions. It has been shown that activation of PPAR by synthetic ligands or components of oxidized LDL reduces monocyte CCR2 expression and blocks chemotaxis mediated by MCP-1 (30). In parallel, activation of PPAR by pioglitazone may be responsible for the observed effects in our study. While no human data has been published yet about the inuence of pioglitazone on plasma MCP-1 levels, recent animal experiments have indicated and demonstrated the prevention of coronary arteriosclerosis by additional MCP-1related antiinammatory effects (down-regulation of CCR2 in circulating and lesional monocytes) (16,31). In a small study without an active comparator, six weeks of treatment with 4 mg of rosiglitazone resulted in a signicant improvement of plasma MCP-1 in diabetic and nondiabetic subjects (17). While no data have been published about the inuence of pioglitazone on plasma MMP-9 yet, two clinical reports describe the treatment effects of rosiglitazone on this marker. In both cases, however, the studies were only placebo-controlled, and signicant differences between the treatment and comparator groups in long-term blood glucose control and HbA1c were the consequence of this design. In one study, MMP-9 decreased in a dosedependent manner by 12.4% (4 mg dose) and 23.4% (8 mg dose) during 26 weeks of therapy as compared to placebo (28). In the other trial, 4 mg of rosiglitazone twice daily led to a signicant reduction in MMP-9 by 24.1% (compared to baseline) after 12 weeks (18). In the presented study, however, the 14.5% decrease in MMP-9 under pioglitazone compares to an increase by 2.8% with a glimepiride treatment that results in the same HbA1c improvement. No signicant changes for sCD40L have been detected in our study. Reduction of this risk marker has been independently reported after treatment with rosiglitazone and troglitazone. A reduction of sCD40 by 18.4% after six weeks of treatment with rosiglitazone (4 mg twice a day) in comparison to placebo was reported by Marx et al. (19), and a mean reduction by 29% in a heterogeneous diabetic population was reported after troglitazone treatment for 12 weeks (12). However, in both study groups, the baseline values of sCD40L were about twice as high as in our study popula-

1930

Pftzner et al. Cardiovascular Risk Improvement by Pioglitazone

JACC Vol. 45, No. 12, 2005 June 21, 2005:192531


6. Willerson JT, Ridker PM. Inammation as cardiovascular risk factor. Circulation 2004;109 Suppl 1:II210. 7. Aukrust P, Muller F, Ueland T, et al. Enhanced levels of soluble and membrane-bound CD40 ligand in patients with unstable angina. Possible reection of T lymphocyte and platelet involvement in the pathogenesis of acute coronary syndromes. Circulation 1999;100:614 20. 8. Cipollone F, Mezzetti A, Porreca E, et al. Association between enhanced soluble CD40L and prothrombotic state in hypercholesterolemia: effects of statin therapy. Circulation 2002;106:399 402. 9. Death AK, Fisher EJ, McGrath KC, Yue DK. High glucose alters matrix metalloproteinase expression in two key vascular cells: potential impact on atherosclerosis in diabetes. Atherosclerosis 2003;168:2639. 10. Ferroni P, Basili S, Martini F, et al. Serum metalloproteinase 9 levels in patients with coronary artery disease: a novel marker of inammation. J Investig Med 2003;51:295300. 11. Blankenberg S, Rupprecht HJ, Poirier O, et al. Plasma concentrations and genetic variation of matrix metalloproteinase 9 and prognosis of patients with cardiovascular disease. Circulation 2003; 107:1579 85. 12. Varo N, Vicent D, Libby P, et al. Elevated plasma levels of the atherogenic mediator soluble CD40 ligand in diabetic patients: a novel target of thiazolidinediones. Circulation 2003;107:2664 9. 13. Pasceri V, Cheng JS, Willerson JT, Yeh ET, Chang J. Modulation of C-reactive protein-mediated monocyte chemoattractant protein-1 induction in human endothelial cells by anti-atherosclerosis drugs. Circulation 2001;103:2531 4. 14. Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inammatory cytokines. Nature 1998;391:82 6. 15. Marx N, Sukhova G, Murphy C, Libby P, Plutzky J. Macrophages in human atheroma contain PPARgamma: differentiation-dependent peroxisomal proliferators-activated receptor gamma (PPARgamma) expression and reduction of MMP-9 activity through PPARgamma activation in mononuclear phagocytes in vitro. Am J Pathol 1998;153: 1723. 16. Ito H, Nakano A, Kinoshita M, Matsumori A. Pioglitazone, a peroxisome proliferator-activated receptor-gamma agonist, attenuates myocardial ischemia/reperfusion injury in a rat model. Lab Invest 2003;83:171521. 17. Mohanty P, Aljada A, Ghanim H, et al. Evidence for a potent antiinammatory effect of rosiglitazone. J Clin Endocrinol Metab 2004;89:2728 35. 18. Marx N, Froehlich J, Siam L, et al. Antidiabetic PPAR gammaactivator rosiglitazone reduces MMP-9 serum levels in type 2 diabetic patients with coronary artery disease. Arterioscler Thromb Vasc Biol 2003;23:283 8. 19. Marx N, Imhof A, Froehlich J, et al. Effect of rosiglitazone treatment on soluble CD40L in patients with type 2 diabetes and coronary artery disease. Circulation 2003;107:1954 7. 20. Tang R, Hennig M, Thomasson B, et al. Baseline reproducibility of B-mode ultrasonic measurement of carotid artery intima-media thickness: the European Lacidipine Study of Atherosclerosis (ELSA). J Hypertens 2000;18:197201. 21. Bots ML, Evans GW, Riley WA, Grobbee DE. Carotid intima-media thickness measurements in intervention studies: design options, progression rates, and sample size considerations: a point of view. Stroke 2003;34:298594. 22. Tan M, Johns D, Gonzalez Galvez G, et al., for the GLAD Study Group. Effects of pioglitazone and glimepiride on glycemic control and insulin sensitivity in Mexican patients with type 2 diabetes mellitus: a multicenter, randomized, double-blind, parallel-group trial. Clin Ther 2004;26:680 93. 23. Tan MH, Johns D, Strand J, et al. Sustained effects of pioglitazone vs. glibenclamide on insulin sensitivity, glycaemic control, and lipid proles in patients with type 2 diabetes. Diabet Med 2004;21:859 66. 24. Derosa G, Cicero AF, Gaddi A, et al. Metabolic effects of pioglitazone and rosiglitazone in patients with diabetes and metabolic syndrome treated with glimepiride: a twelve-month, multicenter, double-blind, randomized, controlled, parallel-group trial. Clin Ther 2004;26:744 54. 25. King AB, Armstrong DU. Lipid response to pioglitazone in diabetic patients: clinical observations from a retrospective chart review. Diabetes Technol Ther 2002;4:14551.

tion, which may explain our inability to observe any significant differences with regard to this parameter. No inuence of pioglitazone could also be observed on PAI-1 levels. Some authors describe reduction of PAI-1 expression by TZDs (32,33), but others found no effect on PAI-1 expression at all (34). All these observations, however, have been made in vitro. Osman et al. (35) measured PAI-1 in type 2 patients with restenosis treated with rosiglitazone and could not nd any changes in their study population (35). The major clinical nding of this study is a signicant reduction of carotid IMT, a strong and well described clinical predictor of cardiovascular risk and stroke (36,37), exclusively in the pioglitazone-treated study population. This nding and the parallel reduction in several biochemical risk markers including hsCRP, MCP-1, MMP-9, and the increase in adiponectin strongly suggest substantial antiarteriosclerotic actions of pioglitazone in vivo independent from metabolic control. While anti-inammatory and antiarteriosclerotic effects of pioglitazone have been suggested after analysis of animal experiments (31), this is a rst comprehensive clinical investigation of these effects in humans. The answer to the question, whether the surrogate ndings described in this study report can be translated into substantial clinical outcome improvements, is currently under investigation in the Prospective Pioglitazone Clinical Trial in Macrovascular Events (PROactive) study with 5,238 patients with type 2 diabetes. The cohort of patients enrolled in PROactive is a typical type 2 diabetic population at high risk of further macrovascular events. The primary end point is the time from randomization to occurrence of a new macrovascular event or death (38). In conclusion, the presented study gives evidence of an anti-inammatory and potential antiatherogenic effect of pioglitazone that is indicated by improvements in several traditional and nontraditional cardiovascular risk markers and carotid IMT, independent of an improvement in long-term glycemic control.
Reprint requests and correspondence: Dr. Andreas Pftzner, Institute for Clinical Research and Development, Parcusstr. 8, D-55116 Mainz, Germany. E-mail: AndreasP@ikfe.de.

REFERENCES
1. Howard B, Rodriguez B, Bennet P, et al. Prevention conference VI: diabetes and cardiovascular disease: writing group I: epidemiology. Circulation 2002;105:e1327. 2. Haffner SM, Lehto S, Ronnemaa T, Pyorala K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998;339:229 34. 3. Ross R. Artherosclerosis: an inammatory disease. N Engl J Med 1999;340:11526. 4. Ridker PM. Evaluating novel cardiovascular risk factors: can we better predict heart attacks? Ann Intern Med 1999;130:9337. 5. Ridker PM, Hennekens CH, Buring JE, Rifai N. C-reactive protein and other markers of inammation in the prediction of cardiovascular disease in women. N Engl J Med 2000;342:836 43.

JACC Vol. 45, No. 12, 2005 June 21, 2005:192531


26. Saad MF, Greco S, Osei K, et al. Ragaglitazar improves glycemic control and lipid prole in type 2 diabetic subjects: a 12-week, double-blind, placebo-controlled dose-ranging study with an open pioglitazone arm. Diabetes Care 2004;27:1324 9. 27. Tordjman J, Chauvet G, Quette J, Beale EG, Forest C, Antoine B. Thiazolidinediones block fatty acid release by inducing glyceroneogenesis in fat cells. J Biol Chem 2003;278:1878590. 28. Haffner SM, Greenberg AS, Weston WM, Chen H, Williams K, Freed MI. Effect of rosiglitazone treatment on nontraditional markers of cardiovascular disease in patients with type 2 diabetes mellitus. Circulation 2002;106:679 84. 29. Satoh N, Ogawa Y, Usui T, et al. Antiatherogenic effect of pioglitazone in type 2 diabetic patients irrespective of the responsiveness to its antidiabetic effect. Diabetes Care 2003;26:24939. 30. Han KH, Quehenberger O. Ligands for peroxisome proliferatoractivated receptor inhibit monocyte CCR2 expression stimulated by plasma lipoproteins. Trends Cardiovasc Med 2000;10:209 16. 31. Ishibashi M, Egashira K, Hiasa K, et al. Anti-inammatory and antiarteriosclerotic effects of pioglitazone. Hypertension 2002;40:68793. 32. Kato K, Satoh H, Endo Y, et al. Thiazolidinediones down-regulate plasminogen activator inhibitor type 1 expression in human vascular endothelial cells: a possible role for PPARgamma in endothelial function. Biochem Biophys Res Commun 1999;258:4315.

Pftzner et al. Cardiovascular Risk Improvement by Pioglitazone

1931

33. Zirlik A, Leugers A, Lohrmann J, et al. Direct attenuation of plasminogen activator inhibitor type-1 expression in human adipose tissue by thiazolidinediones. Thromb Haemost 2004;91:674 82. 34. He G, Pedersen SB, Bruun JM, Lihn AS, Richelsen B. Metformin, but not thiazolidinediones, inhibits plasminogen activator inhibitor-1 production in human adipose tissue in vitro. Horm Metab Res 2003;35:18 23. 35. Osman A, Otero J, Brizolara A, et al. Effect of rosiglitazone on restenosis after coronary stenting in patients with type 2 diabetes. Am Heart J 2004;147:e23. 36. Bots ML, Hoes AW, Koudstaal PJ, Hofman A, Grobbee DE. Common carotid intima-media thickness and risk of stroke and myocardial infarction: the Rotterdam study. Circulation 1997;96: 14327. 37. Touboul PJ, Elbaz A, Koller C, et al. Common carotid artery intima-media thickness and brain infarction: the Etude du Prol Genetique de lInfarctus Cerebral (GENIC) case-control study. The GENIC Investigators. Circulation 2000;102:313 8. 38. Charbonnel B, Dormandy J, Erdmann E, Massi-Benedetti M, Skene A, PROactive Study Group. The prospective pioglitazone clinical trial in macrovascular events (PROactive): can pioglitazone reduce cardiovascular events in diabetes? Study design and baseline characteristics of 5,238 patients. Diabetes Care 2004;27:164753.

You might also like