Skip to main content
Advertisement
  • Loading metrics

The Effect of Sitagliptin on Carotid Artery Atherosclerosis in Type 2 Diabetes: The PROLOGUE Randomized Controlled Trial

Abstract

Background

Experimental studies have suggested that dipeptidyl peptidase-4 (DPP-4) inhibitors provide cardiovascular protective effects. We performed a randomized study to evaluate the effects of sitagliptin added on to the conventional therapy compared with conventional therapy alone (diet, exercise, and/or drugs, except for incretin-related agents) on the intima-media thickness (IMT) of the carotid artery, a surrogate marker for the evaluation of atherosclerotic cardiovascular disease, in people with type 2 diabetes mellitus (T2DM).

Methods and Findings

We used a multicenter PROBE (prospective, randomized, open label, blinded endpoint) design. Individuals aged ≥30 y with T2DM (6.2% ≤ HbA1c < 9.4%) were randomly allocated to receive either sitagliptin (25 to 100 mg/d) or conventional therapy. Carotid ultrasound was performed at participating medical centers, and all parameters were measured in a core laboratory. Of the 463 enrolled participants with T2DM, 442 were included in the primary analysis (sitagliptin group, 222; conventional therapy group, 220). Estimated mean (± standard error) common carotid artery IMT at 24 mo of follow-up in the sitagliptin and conventional therapy groups was 0.827 ± 0.007 mm and 0.837 ± 0.007 mm, respectively, with a mean difference of −0.009 mm (97.2% CI −0.028 to 0.011, p = 0.309). HbA1c level at 24 mo was significantly lower with sitagliptin than with conventional therapy (6.56% ± 0.05% versus 6.72% ± 0.05%, p = 0.008; group mean difference −0.159, 95% CI −0.278 to −0.041). Episodes of serious hypoglycemia were recorded only in the conventional therapy group, and the rate of other adverse events was not different between the two groups. As it was not a placebo-controlled trial and carotid IMT was measured as a surrogate marker of atherosclerosis, there were some limitations of interpretation.

Conclusions

In the PROLOGUE study, there was no evidence that treatment with sitagliptin had an additional effect on the progression of carotid IMT in participants with T2DM beyond that achieved with conventional treatment.

Trial Registration

University Hospital Medical Information Network Clinical Trials Registry UMIN000004490

Author Summary

Why Was This Study Done?

  • Translational research suggests that dipeptidyl peptidase-4 (DPP-4) inhibitors, which are drugs for type 2 diabetes mellitus (T2DM), may prevent cardiovascular disease to a greater extent than other T2DM drugs. However, recently published large clinical trials showed no additional efficacy for DPP-4 inhibitors beyond that expected for the drugs’ glycemic control. Possibly these studies showed no benefit to cardiovascular health because they assessed cardiovascular mortality and other relatively rare outcomes.
  • Change in carotid intima-media thickness (IMT) is a simple, noninvasive method to assess atherosclerosis, an intermediate and more common condition that can precede cardiovascular disease.

What Did the Researchers Do and Find?

  • We conducted a trial to evaluate whether DPP-4 inhibitors affect atherosclerosis in people with T2DM. In all, 463 participants recruited at 48 medical centers were randomly divided into two groups and treated with sitagliptin, a DPP-4 inhibitor, or other conventional drugs for two years. Carotid IMT was evaluated with ultrasonography in a blinded manner.
  • We found that annual changes in carotid IMT were not significantly different between the two groups. On the other hand, the decrease in HbA1c and in the incidence of hypoglycemia with sitagliptin were superior to conventional therapy. The rate of other adverse events was not different between the two groups.

What Do These Findings Mean?

  • These findings provide no evidence that sitagliptin slows the progression of carotid intima-media thickening in participants with T2DM to a greater extent than conventional drugs. As some of the conventional anti-T2DM drugs have shown anti-atherosclerotic effects in clinical trials, the present findings do not mean that sitagliptin does not possess anti-atherosclerotic properties in participants with T2DM.
  • Importantly, however, sitagliptin is useful for controlling hyperglycemia for clinical setting.

Introduction

Atherosclerosis, often caused by hypertension, diabetes mellitus, or dyslipidemia, causes ischemic diseases of the brain, heart, and kidney, which increase mortality and morbidity worldwide. Above all, diabetes mellitus is a critical factor in the development of vascular injury. The global prevalence of type 2 diabetes mellitus (T2DM) has been estimated to be 171 million cases, and it is projected to reach double this estimate or more by 2030 [1]. Epidemiological studies have shown that the mortality caused by T2DM is equivalent to that resulting from coronary artery disease (CAD) [24].

Noninvasive assessment of carotid artery intima-media thickness (IMT) is widely used to estimate not only carotid but also systemic atherosclerosis, because carotid IMT is independently associated with both the presence of CAD [58] and the occurrence of cardiovascular events [9,10], including after adjustment for conventional cardiovascular risk factors. It has also been suggested that an increased carotid IMT is strongly associated with the presence of CAD in people with T2DM [11]. Therefore, evaluation of carotid IMT is helpful when treating people with T2DM.

Dipeptidyl peptidase-4 (DPP-4) inhibitors are antihyperglycemic drugs that stimulate insulin release from pancreatic β-cells by sparing incretin hormones, such as glucagon-like peptide-1 (GLP-1), from degradation by DPP-4. GLP-1 analogues and DPP-4 inhibitors have also been shown to have extra-pancreatic effects beyond glycemic control, including anti-atherosclerotic properties [12,13]. Previous meta-analyses of DPP-4 inhibitors in cardiovascular diseases have demonstrated decreases in cardiovascular events [14,15], while large-scale clinical trials have found that DPP-4 inhibitors neither increased nor decreased the onset of cardiovascular events [1618]. The SAVOR-TIMI 53 trial reported a significant 27% increase in the incidence of hospitalization for heart failure [16], indicating that it is unclear whether DPP-4 inhibitors have a cardioprotective effect in the clinical setting. Moreover, the anti-atherosclerotic effects of DPP-4 inhibitors are not yet fully clarified. We designed the PROLOGUE (Program of Vascular Evaluation under Glucose Control by DPP-4 Inhibitor) study to investigate the effect of sitagliptin on carotid IMT in people with T2DM.

Methods

Study Design and Participants

The rationale and design of the PROLOGUE study have already been described [19]. The study was a multicenter PROBE (prospective, randomized, open label, blinded endpoint) trial carried out in 48 Japanese medical centers. The study protocol and all amendments were reviewed and approved by each center’s ethics committee (S1 and S2 Texts). The study was conducted in accordance with the ethical principles of the Declaration of Helsinki and conformed with good clinical practices and applicable regulatory requirements. All participants gave written consent to participate after having been informed about the nature and purpose of the study, the participation/termination conditions, and the possible risks and benefits of treatment.

We enrolled 463 people with T2DM between November 2010 and September 2012. The inclusion criteria were age ≥ 30 y and presence of T2DM with HbA1c ≥ 6.2% and < 9.4% despite treatment with diet, exercise, and/or conventional antidiabetic agents. The exclusion criteria were as follows: type 1 diabetes mellitus; undergoing insulin treatment; administration of DPP-4 inhibitors and/or GLP-1 analogues before randomization; heart failure with New York Heart Association functional class III or IV; a history of diabetic ketoacidosis or diabetic coma within the 6 mo prior to randomization; a history of myocardial infarction, angina pectoris, percutaneous transluminal coronary angioplasty, or bypass surgery; a history of cerebral infarction, cerebral hemorrhage, subarachnoid hemorrhage, or transient ischemic attack within the 3 mo prior to randomization; serious renal dysfunction (estimated glomerular filtration rate < 30 ml/min/1.73 m2 or dialysis); pregnancy or possible pregnancy; lack of informed consent; and judgment of the investigator that an individual is ineligible for inclusion in the study.

Following the initial echocardiographic estimation of carotid IMT, eligible participants were randomly assigned (ratio 1:1) either to receive conventional therapy plus sitagliptin (sitagliptin group) or to continue with only conventional therapy (diet, exercise, and/or antidiabetic agents, except for DPP-4 inhibitors, GLP-1 analogues, and insulin; conventional therapy group). The randomization was done at the PROLOGUE data center using a modified minimization method with a biased-coin assignment balancing on age (<65 or ≥65 y), sex, use of statins, use of antidiabetic agents (nonpharmacological or pharmacological), HbA1c (<7.0% or ≥7.0%), office systolic blood pressure (<135 or ≥135 mm Hg), and maximum IMT (<1.0 or ≥1.0 mm). The random allocation with a stratified technique was generated automatically by a centralized web-based tool that could not be influenced by the researchers. The treatment target level of HbA1c in both groups was <6.2%. All participants were followed annually for 2 y until September 2014 (S1 Fig).

Measurement of Carotid Intima-Media Thickness

Carotid ultrasound examinations were performed within 1 mo prior to study inclusion, and at 12 and 24 mo after randomization. High-resolution carotid ultrasonography was performed in each ultrasound laboratory in a blinded manner using standardized imaging protocols by expert sonographers trained in the measurement of carotid IMT [20]. All ultrasound systems were equipped with linear transducers of more than 7.5 MHz. Longitudinal B-mode images, perpendicular to the ultrasound beam, with a 4-cm imaging depth, were obtained from the distal common carotid arteries (CCAs), bulbs, and proximal internal carotid arteries (ICA) on both sides. The lateral probe incidence was used to obtain CCA images, using external landmarks with an original semicircular protractor developed for the purpose. The IMT primary parameter measured and calculated was change in the mean far wall CCA IMT in the right and left CCAs 10 mm from the bulb. In addition, the following far wall IMTs were measured: the maximum IMT of the CCA, the mean of the mean IMTs of the CCA, bulb, and ICA, and the mean of the maximum IMTs of the CCA, bulb, and ICA. We used the method recommended by the Mannheim carotid IMT consensus [21]. Plaque was defined as the presence of focal wall thickening at least 50% greater than that of the surrounding vessel wall, or as a focal region with IMT > 1.1 mm protruding into the lumen and distinct from the adjacent boundary. The plaque gray scale median [22] and plaque area far and near to the wall were also measured. If there were multiple carotid plaques in participants, the most echolucent one was selected for measurement of plaque area and gray scale value. The optimized R-wave-gated still frames of the carotid IMT were stored as JPEG files, and all parameters were measured in a core laboratory (University of Tsukuba). A single expert analyst who was unaware of the clinical information of the participants measured all the IMT values using an automatic IMT measurement software program in a blinded manner (Vascular Research Tools 5, Medical Imaging Applications) [23]. The analyst selected the best images of the right and left CCA, bulb, and ICA. In this region, the software identified the lumen/intima and the media/adventitia borders, and calculated the distance between them.

Endpoints

The primary endpoint was the percentage change in mean CCA IMT at 24 mo after randomization. The secondary endpoints, at 12 and 24 mo, were (1) the mean and maximum IMT values and changes at the CCA, bulb, and ICA (except for the primary endpoint); (2) plaque area and plaque gray scale median; (3) the values and changes in glycemic profiles (HbA1c, fasting glucose level, insulin concentration, 1,5-anhydroglucitol,1,4-anhydro-D-glucitol, HOMA-β, HOMA-R); lipoprotein profiles (total cholesterol, high-density lipoprotein cholesterol, triglyceride, small dense low-density lipoprotein, malondialdehyde-modified low-density lipoprotein, remnant-like particle cholesterol); renal function (creatinine, cystatin C, urinary albumin/creatinine ratio, estimated glomerular filtration rate); high molecular weight adiponectin; physiological parameters (body weight, blood pressure); and (4) adjudicated clinical events and adverse events.

Statistical Analysis

Based on previous reports, we assumed that the annual change in mean CCA IMT would be −0.005 mm/y in the sitagliptin group and +0.005 mm/y in the conventional therapy group [24,25]. The necessary sample size was calculated on the assumption of a 0.01-mm group difference in the primary endpoint at the final assessment, with a standard deviation (SD) of 0.06 for individual differences and 80% power for a two-sided, two-sample t-test at a 0.05 significance level. The target sample size was set at 567 participants per group in anticipation of a 20% dropout rate, with total sample size at 1,200 participants. However, as there has been increasing evidence that DPP-4 inhibitors can reduce HbA1c safely and adequately, they have been quite widely prescribed in Japan. As a consequence, it was difficult to recruit participants who were not taking them, and also to continue the trial without prescribing them. Based on these circumstances, we recalculated the necessary sample size assuming that DPP-4 inhibitors have protective effects similar to those of thiazolidine derivatives, which were reported to prevent carotid intima-media thickening in the CHICAGO study [26]. In the sample size recalculation, we assumed a 0.02-mm group difference with an SD of 0.06 for individual differences and 80% power for a two-sided, two-sample t-test at a 0.05 significance level. The target sample size was re-estimated at 238 participants per group in anticipation of a 20% dropout rate, and the total sample size was set at 500 in June 2012. An interim analysis was done for the purpose of investigating futility or overwhelming efficacy of the study and was performed by an independent data monitoring committee at 1 y after the completion of enrollment. The investigators and participants remained blinded to the results of this interim analysis. Significance was evaluated using the Pocock method (with the stopping boundary p ≤ 0.0311 at the first interim analysis), resulting in approval of study continuation.

The data for the primary and secondary endpoints were collected at each time point. The statistical analysis and reporting of this trial were conducted in accordance with the CONSORT guidelines, with the primary analyses based on the intention to treat principle (S3 Text). For the baseline variables, summary statistics were constructed employing frequencies and proportions for categorical data and mean and SD for continuous variables. The baseline variables were compared using Fisher’s exact test for categorical outcomes and unpaired t-tests for continuous variables, as appropriate.

For the primary analysis comparing the treatment effects, the baseline-adjusted means and their 97.2% CIs, estimated by analysis of covariance (ANCOVA), for the change in average carotid IMT at 24 mo were compared between the treatments (sitagliptin group versus conventional therapy group). This analysis was carried out taking into account the variation caused by treatment effects, and using age, sex, statin use, prerandomization treatment type, baseline HbA1c, baseline office systolic blood pressure, and baseline maximum IMT as covariates. The significance level of the final analysis was set at 0.0277 (two-sided). We did not impute missing observations for the primary analysis, but the mixed-effects model for repeated measures was applied as a sensitivity analysis to examine the effect of missing data. The secondary analyses were performed in the same manner as the primary analysis. The baseline-adjusted means of the secondary endpoints at each time point were estimated by ANCOVA with treatment effects, age, sex, statin usage, pre-randomization treatment type, baseline HbA1c, baseline office systolic blood pressure, and the baseline value of the secondary efficacy parameter as covariates.

All comparisons were planned, and all p-values were two-sided. A p-value < 0.05 was considered statistically significant. All statistical analyses were performed using SAS, version 9.4 (SAS Institute).

Study Management

The organization of the study is detailed in S4 Text. A steering committee was responsible for the study design and scientific execution. An independent efficacy and safety evaluation committee, consisting of six members blinded to any information related to group allocation, evaluated each clinical and adverse event. The independent data monitoring committee, composed of three members, independently reviewed the interim analysis to advise whether it was appropriate to continue the study. An independent audit team inspected several main medical centers to ensure the quality of the data. All medical centers participating in this study are listed in S4 Text. Changes in the protocol, including definition of the primary endpoint, change of sample size, and enforcement of interim analysis, are described in S2 Text.

Data for this study were deposited in the Dryad Digital Repository [27].

Results

Study Population

In all, 463 people with T2DM were enrolled and randomly assigned to the two groups (232 and 231 to the sitagliptin and conventional therapy groups, respectively). We obtained complete endpoint information at the end of the study for 222 participants in the sitagliptin group and 220 participants in the conventional therapy group (Fig 1). There were no significant differences in baseline characteristics between the two groups (Table 1).

thumbnail
Table 1. Baseline demographics and clinical characteristics.

https://doi.org/10.1371/journal.pmed.1002051.t001

Carotid Intima-Media Thickness

At baseline, the mean CCA IMT (± SD) was 0.829 ± 0.166 mm in the sitagliptin group and 0.835 ± 0.190 mm in the conventional therapy group (Table 1). The baseline-adjusted mean (± standard error [SE]) CCA IMT at 24 mo in the sitagliptin and conventional therapy groups was 0.827 ± 0.007 mm and 0.837 ± 0.007 mm, respectively, with a mean difference of −0.009 mm (97.2% CI −0.028 to 0.011, p = 0.309). Although the change in maximum ICA IMT at 24 mo was significantly greater in the sitagliptin group (p = 0.038), the values of mean bulb IMT, mean ICA IMT, maximum CCA IMT, maximum bulb IMT, plaque area, and plaque gray scale median at 24 mo did not differ significantly between the two groups at any time point (Table 2).

thumbnail
Table 2. IMT measurement, plaque area, and plaque gray scale median at each time point.

https://doi.org/10.1371/journal.pmed.1002051.t002

The effect of sitagliptin on carotid IMT in older participants (≥70 y) was evaluated (S1 Table). However, there was no statistically significant difference in effect between the older and younger participants. Accordingly, the results did not depend on the age of the participants.

Glucose Metabolism and Other Parameters

The baseline levels of HbA1c were 6.96% ± 0.64% and 6.96% ± 0.55% in the sitagliptin and conventional therapy groups, respectively (mean ± SD; Table 1). The baseline-adjusted mean (± SE) at each time point and the change in HbA1c level are shown in Tables S2 and 3. HbA1c level decreased to 6.56% ± 0.04% in the sitagliptin group and to 6.67% ± 0.04% in the conventional therapy group at 12 mo (p = 0.047), and to 6.56% ± 0.05% in the sitagliptin group and 6.72% ± 0.05% in the conventional therapy group at 24 mo (p = 0.008). While the change in 1,5-anhydroglucitol,1,4-anhydro-D-glucitol at 24 mo in the conventional therapy group was significantly greater than that in the sitagliptin group (p = 0.048), there was no significant difference between the groups in fasting plasma glucose at 24 mo (p = 0.787). There were no significant differences between the two groups in changes in the other parameters, including body weight, blood pressure, renal function, and metabolic and lipid profiles.

thumbnail
Table 3. Physiological and biochemical parameters at 24 mo.

https://doi.org/10.1371/journal.pmed.1002051.t003

In the conventional therapy group, the additional use of a sulfonylurea, biguanide, α-glucosidase inhibitor, or thiazolidinedione for the management of glycemic control increased during the 24-mo observation period. Conversely, the usage of other antidiabetic agents, except for biguanide drugs, did not increase in the sitagliptin group (Table 4). The average dose of sitagliptin was 48 ± 8 mg at baseline, 62 ± 24 mg at 12 mo, and 64 ± 26 mg at 24 mo (S3 Table).

In each participant, adherence to medications was monitored by counting the residual number of tablets at each clinic visit. If the average daily consumption of all tablets combined in a participant was less than 75% of that prescribed, the participant was defined as having “poor adherence” at the end of the study. There were seven such participants in the sitagliptin group, and nine in the conventional therapy group.

Adjudicated Clinical and Adverse Events

During the 24 mo of follow-up, five participants died, three in the sitagliptin group and two in the conventional therapy group (nontuberculous mycobacterial infection, multiple organ failure following bone fracture, coma of unknown cause, or sudden death); sudden death occurred in one participant in each group. Nonfatal myocardial infarction occurred in two participants in the sitagliptin group and in one in the conventional therapy group. Nonfatal stroke occurred in one participant in each group. Two participants in each group initiated insulin therapy. Deterioration of renal function, defined as a greater than 2-fold increase in serum creatinine level, was seen in three participants in the sitagliptin group and in one in the conventional therapy group. Hypoglycemia, which is one of the most clinically important adverse events, was recorded only in the conventional therapy group. Heart failure occurred in two participants in the sitagliptin group and in four in the conventional therapy group. Acute pancreatitis and pancreatic cancer were not observed in either group (Table 5).

thumbnail
Table 5. Summary of adjudicated clinical events and adverse events.

https://doi.org/10.1371/journal.pmed.1002051.t005

Discussion

Main Findings

To our knowledge, the PROLOGUE study is the largest trial to investigate whether DPP-4 inhibitors slow the progression of carotid IMT in participants with T2DM. The major finding of the study is that the serial change in mean CCA IMT was not significantly different between conventional treatment plus sitagliptin and conventional treatment alone. The change in HbA1c was significantly greater in the sitagliptin group. These results suggest that sitagliptin failed to inhibit IMT progression relative to conventional therapy despite its glucose-lowering effect.

Intima-Media Thickness in People with Type 2 Diabetes Mellitus

The primary endpoint of carotid IMT in this study, which can be evaluated noninvasively using ultrasonography, reflects atherosclerotic status and has been used to predict the risk of myocardial infarction and stroke [28]. A positive association exists between carotid IMT and subsequent cardiovascular events in the general population, independent of other major risk factors [29]. A single measurement of carotid IMT and assessment of carotid plaque increases the predictive power of cardiovascular disease risk assessment [3035]. Accordingly, the American College of Cardiology Foundation/American Heart Association guidelines [36] give carotid IMT and plaque measurement for cardiovascular risk stratification as a class IIA recommendation. Repeated carotid IMT measurements are an accepted approach to test the effects of interventions on carotid IMT progression. The Multi-Ethnic Study of Atherosclerosis [31] showed a positive association between carotid IMT progression and stroke incidence. Although two meta-regression analyses of clinical trial data [37,38] reported conflicting results on the relationship between carotid IMT progression and cardiovascular events, many reports have supported its use in pathophysiological studies and clinical trials, such that the perception of carotid IMT has shifted from that of a secondary endpoint to that of a surrogate marker of the risk of cardiovascular events for evaluating therapeutic interventions in atherosclerotic disease [39,40]. It was on this basis that we undertook the PROLOGUE study to determine whether treatment with a DPP-4 inhibitor, sitagliptin, is more effective than conventional therapy at inhibiting atherosclerotic progression in Japanese people, using carotid IMT as a surrogate marker. Measurement of carotid IMT in a blinded manner and in a core laboratory—as done in this study—avoids bias, improves accuracy, enhances the credibility of image assessments, and ensures consistency. A highly standardized imaging acquisition protocol in a fully blinded manner is recommended [41].

In the CHICAGO (Carotid Intima-Media Thickness in Atherosclerosis Using Pioglitazone) trial, the inhibitory effect of 15–45 mg/d of pioglitazone on mean CCA IMT progression was evident during 18 mo of observation, in comparison with 1–4 mg/d glimepiride [26]. Other investigators also demonstrated that pioglitazone slowed the progression of carotid IMT compared with glimepiride [35,42]. In addition to pioglitazone, a biguanide that has been suggested to inhibit DPP-4 activity and increase GLP-1 concentration [43,44] also reduced the progression of carotid IMT [45,46]. In studies of small numbers of people, DPP-4 inhibitors including sitagliptin were reported to inhibit the progression of carotid IMT [47,48]. The SPEAD-A study revealed that treatment with alogliptin, the other DPP-4 inhibitor, attenuated the progression of carotid IMT compared with conventional treatment during 24 mo of follow-up [49]. More recently, the SPIKE trial demonstrated that sitagliptin attenuated carotid IMT in people with T2DM who were under treatment with insulin [50]. However, compared with these studies showing positive results, in the PROLOGUE study the change in mean CCA IMT in the sitagliptin group did not differ significantly from that in the conventional therapy group. Participants on insulin treatment were excluded from the present study, HbA1c values at baseline were lower (PROLOGUE study: 6.96% [sitagliptin], 6.96% [conventional]; SPEAD-A study: 7.3% [alogliptin], 7.2% [conventional]; SPIKE trial: 8.1% [sitagliptin], 8.0% [conventional]), and the difference between the treatment groups in the change in HbA1c over 24 mo was smaller (PROLOGUE study: 0.14%; SPEAD-A study: 0.2%; SPIKE trial: 0.3%). In addition, the prevalence of statin use was higher in the PROLOGUE study than in the SPEAD-A study and the SPIKE trial (PROLOGUE study: 76.1% [sitagliptin], 73.6% [conventional]; SPEAD-A study: 38% [alogliptin], 46% [conventional]; SPIKE trial: 48% [sitagliptin], 46% [conventional]) [49,50]. Thus, it may not be appropriate to directly compare these different results, as the baseline characteristics of the participants, such as severity of T2DM, HbA1c level, and carotid IMT, differed between them. In the PROLOGUE study, even in the conventional therapy group mean CCA IMT had increased only slightly at 24 mo, which seems to be lower than values reported previously. It has been reported that the annual progression of carotid IMT in Japanese people with T2DM is 0.016 to 0.035 mm [25,51] even with aggressive antidiabetic treatment. In the CHICAGO study, the glimepiride arm showed a 0.012-mm increase in mean CCA IMT during 18 mo of follow-up. Interestingly, in the PROLOGUE study additional use of both pioglitazone and biguanides increased in the conventional therapy group, possibly contributing to the suppression of IMT progression. Additionally, both the sitagliptin and conventional treatment groups achieved strict overall management of risk factors, including glycemic control, although the reduction of HbA1c was greater in the sitagliptin group than in the conventional therapy group. On these grounds, the progression of carotid IMT might be inhibited in both groups, in contrast to previous reports [25,26,4751].

DPP-4 Inhibitors and Cardiovascular Disease

There are published experimental data showing that incretin therapy, including treatment with GLP-1 analogues or DPP-4 inhibitors, attenuates atherosclerosis. GLP-1 and its analogues inhibit the inflammatory and atherogenic pathways induced by exogenous stimulation with oxidative stress and/or inflammation in vitro [5255]. In vivo studies have revealed that the administration of GLP-1 analogues or DPP-4 inhibitors reduces high-fat-diet-induced atherosclerosis and stabilizes plaques in apoE knockout mice [5557]. In the clinical setting, several investigators have demonstrated that GLP-1 analogues or DPP-4 inhibitors improve endothelial function in people with T2DM [58,59]. These lines of evidence suggest that sitagliptin may have beneficial effects on atherosclerosis in humans. Large-scale randomized clinical trials found that DPP-4 inhibitors were safe and effective in the treatment of T2DM. However, they did not reduce cardiovascular events in participants with T2DM who were at high risk for cardiovascular disease or who had a recent onset of acute coronary syndrome [1618]. In the TECOS trial [18], sitagliptin had no effect on cardiovascular events in participants with T2DM. The discrepancy between the experimental data and recent clinical results requires explanation. One possibility is that the period of observation in clinical studies may have been too short to evaluate the clinical effects of sitagliptin. Efficacy against cardiovascular events can be demonstrated over several years, according to the VADT study and the UK Prospective Diabetes Study. The TECOS trial was designed to show noninferiority for cardiovascular events with sitagliptin. Therefore, we had to clarify the precise effects of sitagliptin on atherosclerosis directly to evaluate carotid IMT. In the PROLOGUE study, intima-media thickening did not differ significantly between the sitagliptin and conventional therapy groups during 24 mo of follow-up.

In experimental research in patients with T2DM, comparisons of drugs are relatively simple and their doses are sometimes much greater than those used in clinical practice. Usually people are also taking other drugs, including antidiabetic, antihypertensive, or lipid-lowering drugs, and it is difficult to exclude their influence. Indeed, the use of thiazolidinediones and biguanides increased as co-therapies in the conventional therapy group in our study (Table 4), which may have diminished the effect of the DPP-4 inhibitor. Moreover, better glycemic control can now be achieved compared with several years ago. This may be the reason why carotid IMT in the conventional therapy group did not increase as much as anticipated. We emphasize that the true purpose of the present study was not to elucidate whether sitagliptin can prevent carotid IMT compared with a placebo, but to investigate whether sitagliptin can confer an additional beneficial effect on carotid IMT in clinical practice.

Study Limitations

The PROLOGUE study has a few limitations. First, it was conducted with a PROBE design, which might have introduced bias in the assessment of outcomes. Although the primary endpoint was measured by a single observer in a core laboratory who was blinded to the treatment assignments, open-label treatments might affect physicians’ choices of therapy. Second, baseline HbA1c level and carotid IMT were not very high—probably because participants’ diabetes was well-controlled without insulin treatment—in comparison with previous clinical studies that assessed the additional effects of sitagliptin on carotid atherosclerosis relative to conventional antidiabetic drugs [50].

In conclusion, the PROLOGUE study did not provide evidence that treatment with sitagliptin has an additional effect on the progression of carotid IMT in participants with T2DM beyond that achieved with conventional treatment.

Supporting Information

S1 Fig. Study protocol.

Inclusion criteria: age ≥ 30 y, T2DM with HbA1c ≥ 6.2% and < 9.4%. Exclusion criteria: type 1 diabetes mellitus; receiving insulin therapy; administration of DPP-4 inhibitors or GLP-1 analogues before randomization; heart failure graded at New York Heart Association functional class III or IV; a history of diabetic ketoacidosis or diabetic coma within the 6 mo prior to randomization; a history of myocardial infarction, angina pectoris, percutaneous transluminal coronary angioplasty, or bypass surgery; a history of cerebral infarction, cerebral hemorrhage, subarachnoid hemorrhage, or transient ischemic attack within the 3 mo prior to randomization, serious renal dysfunction (estimated glomerular filtration rate < 30 ml/min/1.73 m2 or dialysis); pregnancy or possible pregnancy; lack of informed consent; and judgment of the investigator that an individual is ineligible for inclusion in the study.

https://doi.org/10.1371/journal.pmed.1002051.s001

(TIF)

S1 Table. The effect of sitagliptin on carotid IMT in younger (<70 y old) and older (≥70 y old) participants.

https://doi.org/10.1371/journal.pmed.1002051.s002

(DOCX)

S2 Table. Physiological and biochemical parameters at 12 mo.

https://doi.org/10.1371/journal.pmed.1002051.s003

(DOCX)

S3 Table. Doses of sitagliptin during the study, Data are presented as the number of participants (percent).

https://doi.org/10.1371/journal.pmed.1002051.s004

(DOCX)

S6 Text. Full competing interests statement.

https://doi.org/10.1371/journal.pmed.1002051.s010

(DOCX)

Acknowledgments

The authors gratefully acknowledge the technical assistance of Sae Katafuchi. The authors thank Akira Yamashina, Takanori Yasu, and Mitsuyoshi Urashima for their key role as the members of the independent data monitoring committee.

PROLOGUE Study Investigators

The PROLOGUE study is a multicenter collaboration. In addition to the listed authors, the following PROLOGUE Study Investigators were involved in this study: Toru Aoyama (Cardiology Center, Nagoya Kyoritsu Hospital); Tetsuya Babazono (Department of Medicine, Diabetes Center, Tokyo Women’s Medical University School of Medicine); Hiroyuki Daida (Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine); Jun Fukui (Division of Cardiology, Hokusho Central Hospital); Kumiko Hamano (Department of Diabetes and Endocrinology, Kanto Rosai Hospital); Shigemasa Hashimoto (Department of Cardiology, Karatsu Red Cross Hospital); Kazunori Hayashi (Department of Cardiology, Nakatsugawa Municipal Hospital); Tsutomu Hirano (Department of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine); Hideki Horibe (Department of Cardiovascular Medicine, Gifu Prefectural Tajimi Hospital); Kazuo Ibaraki (Department of Internal Medicine, Karatsu Red Cross Hospital); Masato Iida (Department of Cardiology, Mitsubishi Nagoya Hospital); Takako Iino (Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine); Kenji Iino (Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine); Yutaka Ishibashi (Department of General Medicine, Shimane University Faculty of Medicine); Yuko S. Ishiguro (Department of Cardiology, Mitsubishi Nagoya Hospital); Ryoji Ishiki (Division of Internal Medicine, Toyota Memorial Hospital); Hiroshi Ito (Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine); Masaaki Ito (Department of Cardiology and Nephrology, Mie University Graduate School of Medicine); Yoshito Iwama (Department of Cardiology, Meijo Hospital); Hideo Izawa (Department of Cardiology, Fujita Health University Banbuntane Hotokukai Hospital); Kenshi Kan (Division of Diabetes, Metabolism and Endocrinology, Tokyo Medical University Hospital); Akira Kimura (Department of Cardiology, Meijo Hospital, Federation of National Public Service Personnel Mutual Aid Association); Ichiro Kishimoto (Department of Atherosclerosis and Diabetes, National Cerebral and Cardiovascular Center); Tomoki Kitano (Department of Cardiology, National Hospital Organization Nagoya Medical Center); Yoshihisa Kizaki (Department of Cardiology, Sasebo Chuo Hospital); Kenji Kohara (Division of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School); Hiroshi Koiwaya (Department of Cardiology, Miyazaki Medical Association Hospital); Taizo Kondo (Department of Cardiology, Gifu Prefectural Tajimi Hospital); Toshimitsu Kosaka (Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine); Nehiro Kuriyama (Department of Cardiology, Miyazaki Medical Association Hospital); Shigetaka Kuroki (Eguchi Hospital); Tatsuya Maruhashi (Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University); Hiroaki Masuzaki (Second Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology, Graduate School of Medicine, University of the Ryukyus); Kaori Miwa (Department of Neurology and Stroke Center, Osaka University Graduate School of Medicine); Takashi Miwa (Department of Diabetes, Endocrinology, Metabolism and Rheumatology, Tokyo Medical University); Tetsuro Miyazaki (Department of Cardiovascular Medicine, Juntendo University School of Medicine); Kazutaka Mori (Department of Cardiology, Nagoya Medical Center); Tomoatsu Mune (Division of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School); Ikue Nakadaira (Diabetes and Endocrinology, Kanto Rosai Hospital); Mashio Nakamura (Department of Cardiology and Nephrology, Mie University Graduate School of Medicine); Yoshihito Nakashima (Department of Cardiovascular Disease, Tosei General Hospital); Masayuki Nakayama (JCHO Saga Central Hospital); Kosaku Nitta (Department of Medicine, Kidney Center, Tokyo Women’s Medical University); Masato Odawara (Department of Diabetes, Endocrinology, Metabolism and Rheumatology, Tokyo Medical University); Yasunori Oguma (Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine); Hirotoshi Ohmura (Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine); Jun Ohno (Department of Cardiology, Tsushima Municipal Hospital); Shinji Okubo (Japan Labour Health and Welfare Organization, Kashima Hospital, Special Department and Cardiology, Tokyo Medical University); Sosho Ri (Division of Diabetes, Metabolism and Endocrinology, Internal Medicine Center, Showa University Koto Toyosu Hospital); Kenji Sadamatsu (Department of Cardiology, Saga Medical Center Koseikan); Makoto Saitoh (Department of Internal Medicine, Nishio Municipal Hospital); Masaki Sakakibara (Department of Cardiology, Handa City Hospital); Hiroaki Sano (Department of Cardiology, Nagoya Ekisaikai Hospital); Yoshisato Shibata (Department of Cardiology, Miyazaki Medical Association Hospital); Toshimasa Shigeta (Department of Cardiology, Gifu Prefectural Tajimi Hospital); Kenei Shimada (Department of Internal Medicine and Cardiology, Osaka City University Graduate School of Medicine); Fuji Somura (Department of Cardiology, Nagoya Central Hospital); Takashi Takei (Department of Medicine, Kidney Center, Tokyo Women’s Medical University); Toshiro Tanaka (Department of Internal Medicine, Nishio Municipal Hospital); Yoshito Tanioka (Division of Cardiology, Omura Municipal Hospital); Akihiro Terasawa (Department of Cardiology, Kasugai Municipal Hospital); Yasuo Terauchi (Department of Endocrinology and Metabolism, Yokohama City University); Masahiko Tsujii (Department of Gastroenterology, Osaka Rosai Hospital); Shuichi Tsuruoka (Department of Nephrology, Nippon Medical School); Hiroyuki Tsutsui (Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine); Hisashi Umeda (Division of Cardiology, Toyota Memorial Hospital); Hiroki Watanabe (Department of Internal Medicine, Nishio Municipal Hospital); Masato Watarai (Cardiovascular Center, Anjo Kosei Hospital); Takaaki Yamada (Department of Cardiology, Nagoya Medical Center); Hiroshi Yamamoto (Cardiovascular Surgery, Yamamoto Memorial Hospital); Chie Yasuoka (Department of Cardiovascular Medicine, Omura Municipal Hospital); Kiyoshi Yokoi (Department of Cardiovascular Medicine, Gifu Prefectural Tajimi Hospital).

Author Contributions

Conceived and designed the experiments: MA YKB KD YH TIn MI TIs KKa HK NK KKi MK KM MM TM MN KN JO MS YS AT HT SU HY KY. Performed the experiments: MA YKB KD YH TIn MI TIs KKa HK NK MK KM MM TM MN KN JO MS HT SU HY KY. Analyzed the data: YS AT. Contributed reagents/materials/analysis tools: TIs YS. Wrote the first draft of the manuscript: MA YKB YH TIn TIs NK MK KM MM TM KN JO MS YS HT SU HY. Contributed to the writing of the manuscript: MA YKB KD YH TIn MI TIs KKa HK NK KKi MK KM MM TM MN KN JO MS YS AT HT SU HY KY. Enrolled patients: MA YB KD YH TIn TIs KKa HK MK KM MM TM MN KN JO HT HY KY. Agree with the manuscript’s results and conclusions: MA YKB KD YH TIn MI TIs KKa HK NK KKi MK KM MM TM MN KN JO MS YS AT HT SU HY KY. All authors are members of the PROLOGUE Study Investigators. All authors have read, and confirm that they meet, ICMJE criteria for authorship.

References

  1. 1. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 2004;27:1047–1053. pmid:15111519
  2. 2. Juutilainen A, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Type 2 diabetes as a “coronary heart disease equivalent”: an 18-year prospective population-based study in Finnish subjects. Diabetes Care. 2005;28:2901–2907. pmid:16306552
  3. 3. Haffner SM, Lehto S, Rönnemaa T, Pyörälä 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;23;339:229–234. pmid:9673301
  4. 4. Whiteley L, Padmanabhan S, Hole D, Isles C. Should diabetes be considered a coronary heart disease risk equivalent?: results from 25 years of follow-up in the Renfrew and Paisley survey. Diabetes Care. 2005;28:1588–1593. pmid:15983305
  5. 5. Craven TE, Ryu JE, Espeland MA, Kahl FR, McKinney WM, Toole JF, et al. Evaluation of the associations between carotid artery atherosclerosis and coronary artery stenosis. A case-control study. Circulation. 1990;82:1230–1242. pmid:2205416
  6. 6. Handa N, Matsumoto M, Maeda H, Hougaku H, Ogawa S, Fukunaga R, et al. Ultrasonic evaluation of early carotid atherosclerosis. Stroke. 1990;21:1567–1572. pmid:2237950
  7. 7. Smith GD, Shipley MJ, Rose G. Intermittent claudication, heart disease risk factors, and mortality. The Whitehall Study. Circulation. 1990;82:1925–1931. pmid:2242518
  8. 8. Khoury Z, Schwartz R, Gottlieb S, Chenzbraun A, Stern S, Keren A. Relation of coronary artery disease to atherosclerotic disease in the aorta, carotid, and femoral arteries evaluated by ultrasound. Am J Cardiol. 1997;80:1429–1433. pmid:9399716
  9. 9. Chambless LE, Heiss G, Folsom AR, Rosamond W, Szklo M, Sharrett AR, et al. Association of coronary heart disease incidence with carotid arterial wall thickness and major risk factors: the Atherosclerosis Risk in Communities (ARIC) Study, 1987–1993. Am J Epidemiol. 1997;146:483–494. pmid:9290509
  10. 10. Salonen JT, Salonen R. Ultrasound B-mode imaging in observational studies of atherosclerotic progression. Circulation. 1993;87:II56–II65. pmid:8443925
  11. 11. Kasami R, Kaneto H, Katakami N, Sumitsuji S, Yamasaki K, Kuroda T, et al. Relationship between carotid intima-media thickness and the presence and extent of coronary stenosis in type 2 diabetic patients with carotid atherosclerosis but without history of coronary artery disease. Diabetes Care. 2011;34:468–470. pmid:21270201
  12. 12. Sivertsen J, Rosenmeier J, Holst JJ, Vilsbøll T. The effect of glucagon-like peptide 1 on cardiovascular risk. Nat Rev Cardiol. 2012;9:209–222. pmid:22290234
  13. 13. Chrysant SG, Chrysant GS. Clinical implications of cardiovascular preventing pleiotropic effects of dipeptidyl peptidase-4 inhibitors. Am J Cardiol. 2012;109:1681–1685. pmid:22425330
  14. 14. Monami M, Ahrén B, Dicembrini I, Mannucci E. Dipeptidyl peptidase-4 inhibitors and cardiovascular risk: a meta-analysis of randomized clinical trials. Diabetes Obes Metab. 2013;15:112–120. pmid:22925682
  15. 15. Patil HR, Al Badarin FJ, Al Shami HA, Bhatti SK, Lavie CJ, Bell DS, et al. Meta-analysis of effect of dipeptidyl peptidase-4 inhibitors on cardiovascular risk in type 2 diabetes mellitus. Am J Cardiol. 2012;110:826–833. pmid:22703861
  16. 16. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med. 2013;369:1317–1326. pmid:23992601
  17. 17. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, et al. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med. 2013;369:1327–1335. pmid:23992602
  18. 18. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, et al. Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2015;373:232–242. pmid:26052984
  19. 19. Oyama J, Ishizu T, Sato Y, Kodama K, Bando YK, Murohara T, et al. Rationale and design of a study to evaluate the effects of sitagliptin on atherosclerosis in patients with diabetes mellitus: PROLOGUE study. Int J Cardiol. 2014;174:383–384. pmid:24768387
  20. 20. Stein JH, Korcarz CE, Hurst RT, Lonn E, Kendall CB, Mohler ER, et al. Use of carotid ultrasound to identify subclinical vascular disease and evaluate cardiovascular disease risk: a consensus statement from the American Society of Echocardiography Carotid Intima-Media Thickness Task Force. Endorsed by the Society for Vascular Medicine. J Am Soc Echocardiogr. 2008;21:93–111. pmid:18261694
  21. 21. Touboul PJ, Hennerici MG, Meairs S, Adams H, Amarenco P, Desvarieux M, et al. Mannheim intima-media thickness consensus. Cerebrovasc Dis. 2004;18:346–349. pmid:15523176
  22. 22. Sabetai MM, Tegos TJ, Nicolaides AN, Dhanjil S, Pare GJ, Stevens JM. Reproducibility of computer-quantified carotid plaque echogenicity: can we overcome the subjectivity? Stroke. 2000;31:2189–2196. pmid:10978050
  23. 23. Lundby-Christensen L, Almdal TP, Carstensen B, Tarnow L, Wiinberg N. Carotid intima-media thickness in individuals with and without type 2 diabetes: a reproducibility study. Cardiovasc Diabetol. 2010;9:40. pmid:20727128
  24. 24. Yamasaki Y, Kodama M, Nishizawa H, Sakamoto K, Matsuhisa M, Kajimoto Y, et al. carotid intima-media thickness in Japanese type 2 diabetic subjects. Diabetes Care. 2000;9:1310–1315.
  25. 25. Kawasumi M, Tanaka Y, Uchino H, Shimizu T, Tamura Y, Sato F, et al. Strict glycemic control ameliorates the increase of carotid IMT in patients with type 2 diabetes. Endocr J. 2006;53:45–50. pmid:16543671
  26. 26. Mazzone T, Meyer PM, Feinstein SB, Davidson MH, Kondos GT, D’Agostino RB Sr, et al. Effect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes. JAMA. 2006;296:2572–2581. pmid:17101640
  27. 27. Oyama J, Node K. Data from: the effect of sitagliptin on carotid artery atherosclerosis in patients with type 2 diabetes: the PROLOGUE randomized controlled trial. Dryad Digital Repository. Avaiable: http://dx.doi.org/10.5061/dryad.qt743/2.
  28. 28. Chambless LE, Folsom AR, Clegg LX, Sharrett AR, Shahar E, Nieto FJ, et al. Carotid wall thickness is predictive of incident clinical stroke: the atherosclerosis risk in communities (ARIC) study. Am J Epidemiol. 2000;151:478–487. pmid:10707916
  29. 29. Lorenz MW, Markus HS, Bots ML, Rosvall M, Sitzer M. Prediction of clinical cardiovascular events with carotid intima-media thickness: a systematic review and meta-analysis. Circulation. 2007;115:459–467. pmid:17242284
  30. 30. Nambi V, Chambless L, Folsom AR, He M, Hu Y, Mosley T, et al. Carotid intima-media thickness and presence or absence of plaque improves prediction of coronary heart disease risk: the ARIC (Atherosclerosis Risk In Communities) study. J Am Coll Cardiol. 2010;55:1600–1607. pmid:20378078
  31. 31. Polak JF, Pencina MJ, O’Leary DH, D’Agostino RB. Common carotid artery intima-media thickness progression as a predictor of stroke in multi-ethnic study of atherosclerosis. Stroke. 2011;42:3017–3021. pmid:21885840
  32. 32. Espeland MA, O’leary DH, Terry JG, Morgan T, Evans G, Mudra H. Carotid intimal-media thickness as a surrogate for cardiovascular disease events in trials of HMG-CoA reductase inhibitors. Curr Control Trials Cardiovasc Med. 2005;6:3. pmid:15760471
  33. 33. Künzli N, Jerrett M, Garcia-Esteban R, Basagaña X, Beckermann B, Gilliland F, et al. Ambient air pollution and the progression of atherosclerosis in adults. PLoS ONE. 2010;5:e9096. pmid:20161713
  34. 34. Hsue PY, Lo JC, Franklin A, Bolger AF, Martin JN, Deeks SG, et al. Progression of atherosclerosis as assessed by carotid intima-media thickness in patients with HIV infection. Circulation. 2004;109:1603–1638. pmid:15023877
  35. 35. Yamasaki Y, Katakami N, Furukado S, Kitagawa K, Nagatsuka K, Kashiwagi A, et al. Long-term effects of pioglitazone on carotid atherosclerosis in Japanese patients with type 2 diabetes without a recent history of macrovascular morbidity. J Atheroscler Thromb. 2010;17:1132–1140. pmid:20686324
  36. 36. Greenland P, Alpert JS, Beller GA, Benjamin EJ, Budoff MJ, Fayad ZA, et al. 2010 ACCF/AHA guideline for assessment of cardiovascular risk in asymptomatic adults: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation. 2010;122:e584–e636. pmid:21098428
  37. 37. Goldberger ZD, Valle JA, Dandekar VK, Chan PS, Ko DT, Nallamothu BK. Are changes in carotid intima-media thickness related to risk of nonfatal myocardial infarction? A critical review and meta-regression analysis. Am Heart J. 2010;160:701–714. pmid:20934565
  38. 38. Costanzo P, Perrone-Filardi P, Vassallo E, Paolillo S, Cesarano P, Brevetti G, et al. Does carotid intima-media thickness regression predict reduction of cardiovascular events? A meta-analysis of 41 randomized trials. J Am Coll Cardiol. 2010;56:2006–2020. pmid:21126642
  39. 39. Amarenco P, Labreuche J, Lavallee P, Touboul PJ. Statins in stroke prevention and carotid atherosclerosis: systematic review and up-to-date meta-analysis. Stroke. 2004;35:2902–2909. pmid:15514180
  40. 40. Riccioni G. Statins and carotid intima-media thickness reduction: an up-to-date review. Curr Med Chem. 2009;16:1799–1805. pmid:19442146
  41. 41. US Food and Drug Administration. Clinical trial imaging endpoint process standards guidance for industry. Draft guidance. 2015 Mar [cited 24 May 2016]. Available: http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm268555.pdf.
  42. 42. Langenfeld MR, Forst T, Hohberg C, Kann P, Lübben G, Konrad T, et al. Pioglitazone decreases carotid intima-media thickness independently of glycemic control in patients with type 2 diabetes mellitus: results from a controlled randomized study. Circulation. 2005;111:2525–2531. pmid:15883215
  43. 43. Green BD, Irwin N, Duffy NA, Gault VA, O’Harte FP, Flatt PR. Inhibition of dipeptidyl peptidase-IV activity by metformin enhances the antidiabetic effects of glucagon-like peptide-1. Eur J Pharmacol. 2005;547:192–199.
  44. 44. Lindsay JR, Duffy NA, McKillop AM, Ardill J, O’Harte FP, Flatt PR, et al. Inhibition of dipeptidyl peptidase IV activity by oral metformin in type 2 diabetes. Diabet Med. 2005;22:654–657. pmid:15842525
  45. 45. Katakami N, Yamasaki Y, Hayaishi-Okano R, Ohtoshi K, Kaneto H, Matsuhisa M, et al. Metformin or gliclazide, rather than glibenclamide, attenuate progression of carotid intima-media thickness in subjects with type 2 diabetes. Diabetologia. 2004;47:1906–1913. pmid:15565373
  46. 46. Matsumoto K, Sera Y, Abe Y, Tominaga T, Yeki Y, Miyake S. Metformin attenuates progression of carotid arterial wall thickness in patients with type 2 diabetes. Diabetes Res Clin Pract. 2004;64:225–258. pmid:15126012
  47. 47. Ishikawa S, Shimano M, Watarai M, Koyasu M, Uchikawa T, Ishii H, et al. Impact of sitagliptin on carotid intima-media thickness in patients with coronary artery disease and impaired glucose tolerance or mild diabetes mellitus. Am J Cardiol. 2014;114:384–388. pmid:24929624
  48. 48. Barbieri M, Rizzo MR, Marfella R, Boccardi V, Esposito A, Pansini A, et al. Decreased carotid atherosclerotic process by control of daily acute glucose fluctuations in diabetic patients treated by DPP-IV inhibitors. Atherosclerosis. 2013;227:349–354. pmid:23375680
  49. 49. Mita T, Katakami N, Yoshii H, Onuma T, Kaneto H, Osonoi T, et al. Alogliptin, a dipeptidyl peptidase 4 inhibitor, prevents the progression of carotid atherosclerosis in patients with type 2 diabetes: the Study of Preventive Effects of Alogliptin on Diabetic Atherosclerosis (SPEAD-A). Diabetes Care. 2016;39:139–148. pmid:26628419
  50. 50. Mita T, Katakami N, Shiraiwa T, Yoshii H, Onuma T, Kuribayashi N, et al. Sitagliptin attenuates the progression of carotid intima-media thickening in insulin-treated patients with type 2 diabetes: the Sitagliptin Preventive Study of Intima-Media Thickness Evaluation (SPIKE): a randomized controlled trial. Diabetes Care. 2016;39:455–464. pmid:26822324
  51. 51. Mita T, Watada H, Ogihara T, Nomiyama T, Ogawa O, Kinoshita J, et al. Eicosapentaenoic acid reduces the progression of carotid intima-media thickness in patients with type 2 diabetes. Atherosclerosis. 2007;191:162–167. pmid:16616147
  52. 52. Shiraki A, Oyama J, Komoda H, Asaka M, Komatsu A, Sakuma M, et al. The glucagon-like peptide 1 analog liraglutide reduces TNF-α-induced oxidative stress and inflammation in endothelial cells. Atherosclerosis. 2012;221:375–382. pmid:22284365
  53. 53. Ishibashi Y, Matsui T, Takeuchi M, Yamagishi S. Glucagon-like peptide-1 (GLP-1) inhibits advanced glycation end product (AGE)-induced up-regulation of VCAM-1 mRNA levels in endothelial cells by suppressing AGE receptor (RAGE) expression. Biochem Biophys Res Commun. 2010;391:1405–1408. pmid:20026306
  54. 54. Liu H, Dear AE, Knudsen LB, Simpson RW. A long-acting glucagon-like peptide-1 analogue attenuates induction of plasminogen activator inhibitor type-1 and vascular adhesion molecules. J Endocrinol. 2009;201:59–66. pmid:19136619
  55. 55. Gaspari T, Liu H, Welungoda I, Hu Y, Widdop RE, Knudsen LB, et al. A GLP-1 receptor agonist liraglutide inhibits endothelial cell dysfunction and vascular adhesion molecule expression in an ApoE-/- mouse model. Diab Vasc Dis Res. 2011;8:117–124. pmid:21562063
  56. 56. Matsubara J, Sugiyama S, Sugamura K, Nakamura T, Fujiwara Y, Akiyama E, et al. A dipeptidyl peptidase-4 inhibitor, des-fluoro-sitagliptin, improves endothelial function and reduces atherosclerotic lesion formation in apolipoprotein E-deficient mice. J Am Coll Cardiol. 2012;59:265–276. pmid:22240132
  57. 57. Vittone F, Liberman A, Vasic D, Ostertag R, Esser M, Walcher D, et al. Sitagliptin reduces plaque macrophage content and stabilises arteriosclerotic lesions in Apoe (-/-) mice. Diabetologia. 2012;55:2267–2275. pmid:22648661
  58. 58. Nyström T, Gutniak MK, Zhang Q, Zhang F, Holst JJ, Ahrén B, et al. Effects of glucagon-like peptide-1 on endothelial function in type 2 diabetes patients with stable coronary artery disease. Am J Physiol Endocrinol Metab. 2004;287:E1209–E1215. pmid:15353407
  59. 59. Kubota Y, Miyamoto M, Takagi G, Ikeda T, Kirinoki-Ichikawa S, Tanaka K, et al. The dipeptidyl peptidase-4 inhibitor sitagliptin improves vascular endothelial function in type 2 diabetes. J Korean Med Sci. 2012;27:1364–1370. pmid:23166419