Skip to main content
Erschienen in: Cardiovascular Diabetology 1/2019

Open Access 01.12.2019 | Original investigation

Sitagliptin does not reduce the risk of cardiovascular death or hospitalization for heart failure following myocardial infarction in patients with diabetes: observations from TECOS

verfasst von: Michael A. Nauck, Darren K. McGuire, Karen S. Pieper, Yuliya Lokhnygina, Timo E. Strandberg, Axel Riefflin, Tuncay Delibasi, Eric D. Peterson, Harvey D. White, Russell Scott, Rury R. Holman

Erschienen in: Cardiovascular Diabetology | Ausgabe 1/2019

Abstract

Background

To examine the effects of the DPP-4i sitagliptin on CV outcomes during and after incident MI in the Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS).

Methods

TECOS randomized 14,671 participants with type 2 diabetes and atherosclerotic cardiovascular disease (ASCVD) to sitagliptin or placebo, in addition to usual care. For those who had a within-trial MI, we analyzed case fatality, and for those with a nonfatal MI, we examined a composite cardiovascular (CV) outcome (CV death or hospitalization for heart failure [hHF]) by treatment group, using Cox proportional hazards models left-censored at the time of the first within-trial MI, without and with adjustment for potential confounders, in intention-to-treat analyses.

Results

During TECOS, 616 participants had ≥ 1 MI (sitagliptin group 300, placebo group 316, HR 0.95, 95% CI 0.81–1.11, P = 0.49), of which 25 were fatal [11 and 14, respectively]). Of the 591 patients with a nonfatal MI, 87 (15%) died subsequently, with 66 (11%) being CV deaths, and 57 (10%) experiencing hHF. The composite outcome occurred in 58 (20.1%; 13.9 per 100 person-years) sitagliptin group participants and 50 (16.6%; 11.7 per 100 person-years) placebo group participants (HR 1.21, 95% CI 0.83–1.77, P = 0.32, adjusted HR 1.23, 95% CI 0.83–1.82, P = 0.31). On-treatment sensitivity analyses also showed no significant between-group differences in post-MI outcomes.

Conclusions

In patients with type 2 diabetes and ASCVD experiencing an MI, sitagliptin did not reduce subsequent risk of CV death or hHF, contrary to expectations derived from preclinical animal models.
Trial registration clinicaltrials.gov no. NCT00790205
Hinweise
Michael A. Nauck and Darren K. McGuire contributed equally to this article

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12933-019-0921-2.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ASCVD
atherosclerotic cardiovascular disease
CV
cardiovascular
DPP-4is
dipeptidyl peptidase-4 inhibitors
hHF
hospitalization for heart failure
MI
myocardial infarction
TECOS
Trial Evaluating Cardiovascular Outcomes with Sitagliptin

Background

Dipeptidyl peptidase-4 inhibitors (DPP-4is) lower plasma glucose and glycated hemoglobin in people with type 2 diabetes by inhibiting degradation of endogenous glucagon-like peptide-1 (GLP-1) [1]. They have a low risk for hypoglycemia and are weight neutral [2]. Although two GLP-1 receptor agonists, once-daily liraglutide [3] and once-weekly semaglutide [4], have been shown to reduce cardiovascular (CV) events in patients with type 2 diabetes at high CV risk, four CV outcome trials that evaluated the once-daily DPP-4i agents saxagliptin [5], alogliptin [6], sitagliptin [7, 8], and linagliptin [9, 10] versus placebo showed no impact on CV death, myocardial infarction (MI), or stroke outcomes.
GLP-1 receptors are expressed on cells in CV tissues [11], and multiple CV effects of GLP-1 receptor agonism have been demonstrated with administration of native GLP-1, with administration of GLP-1 receptor agonists, and with DPP-4i treatment in preclinical studies [1114]. Among these well-documented effects is a substantial (30–50%) reduction in the extent of myocardial necrosis after experimentally induced MI in rodents pretreated with native GLP-1 [15, 16] or with a GLP-1 receptor agonist [17, 18]. Similar experimental approaches with a DPP-4i in mice [19], rats [20], pigs [21], and dogs [22] produced largely similar results. Regarding potential mechanisms, sitagliptin seems to improve tolerance to ischemia as demonstrated by an improved regional contractility in ischemic segments of the left ventricle [23, 24]. These effects of DPP-4 inhibition may be mediated by protection of mitochondrial function and preventing cardiomyocyte apoptosis, and by interfering with oxidative stress during reperfusion [20, 21]. Theoretically, a smaller infarct size in humans could result in lower incident case-fatality, less post-MI arrhythmogenic risk, and higher residual left-ventricular function with a lower future risk of heart failure or CV death [25, 26].
The Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS) randomized patients with type 2 diabetes and atherosclerotic cardiovascular disease (ASCVD) to double-blind therapy with sitagliptin or placebo, in addition to usual care, aiming for glycemic equipoise [7, 8]. In a post hoc analysis, we evaluated the effects of sitagliptin on a composite outcome defined as CV death or hospitalization for heart failure (hHF) in TECOS participants who experienced a within-trial MI.

Methods

Study design

The TECOS design [8] and primary results [7] and heart failure outcomes [27] have been published previously. Briefly, 14,671 participants from 38 countries were enrolled between December 2008 and July 2012. Eligible participants were ≥ 50 years old (no upper age limit) with type 2 diabetes, ASCVD, and glycated hemoglobin (HbA1c) values of 6.5–8.0% (48–64 mmol/mol) on stable dose mono- or dual-combination therapy with metformin, pioglitazone, sulfonylurea or insulin (with or without metformin). Participants were randomized double-blind to sitagliptin or placebo at doses appropriate for their eGFR [7, 8]. During follow-up, treatment for hyperglycemia and for type 2 diabetes comorbidities was provided by usual care providers according to their local guidelines with addition of any open-label glucose-lowering agent permitted, apart from a GLP-1 receptor agonist or DPP-4i. All reported events of death, MI, stroke, and hospitalization for unstable angina or heart failure were adjudicated by an independent committee masked to randomized treatment assignment. Adjudicated event definitions have been published previously [7, 8].

Objectives

The analyses presented here examine only those participants who experienced an MI during the trial. We evaluated potential differences between the randomized groups in case-fatality and for those with a non-fatal MI the time to a composite outcome defined as CV death or hHF. Secondary outcomes were post-MI time to CV death, hHF, and all-cause death. We also examined hHF in patients not known to have heart failure at baseline, and an extended composite outcome defined as CV death, hHF, a further MI, stroke, or new-onset atrial fibrillation.

Statistical analysis

Baseline characteristics for continuous variables were summarized as median and interquartile range (IQR), and categorical variables as count (percentage).
Primary analyses were performed on the intention-to-treat population in the subset who experienced an MI during the trial. Secondary on-treatment sensitivity analyses were performed with participants classified as “DPP-4i treated” if they were taking double-blind sitagliptin study medication or if they were taking an open-label DPP-4i. Similarly, they were classified as “not DPP-4i treated” if they were taking double-blind placebo study medication or had discontinued double-blind sitagliptin study medication and were not taking an open-label DPP-4i.
The two treatment groups were compared using Cox proportional hazards models, without and with adjustment for potential confounders. Adjustment factors applied were those previously identified in the large Nateglinide and Valsartan in Impaired Glucose Tolerance Outcomes Research (NAVIGATOR) clinical trial [28, 29]. The assumptions of linearity and proportional hazards had been previously evaluated for the set of confounders considered and appropriate adjustments applied when violations were noted. The list of covariates is provided in Additional file 1: Table S1. The proportional hazards assumption was tested for the treatment factor in these new models, and time-varying models would have been applied had violations been noted. Follow-up began (day 0) at the date of the first within-trial MI and continued until the date of the first occurrence of each type of endpoint considered here or the date of last contact when no event occurred. The analyses were performed twice in consideration of fatal MIs. In one case (primary analyses), only patients with nonfatal MIs were considered; in the second, the fatal MIs were in the cohort and included as endpoints.
All analyses were performed using SAS version 9.4 (SAS Institute, Cary, NC).

Results

Participant characteristics

Baseline characteristics of all participants at entry to TECOS are listed in Table 1 according to whether or not they had experienced an MI. Those with, compared without, an MI were more likely to be male (77.9% vs. 70.4%, P < 0.0001), to have prior coronary artery disease (89.4% vs. 73.4%, P < 0.0001), prior MI (57.8% vs. 42.0%, P < 0.0001) or prior hHF (21.4% vs. 17.9%, P = 0.024); and to be treated less commonly with metformin (75.5% vs. 81.8%, P < 0.0001) and more commonly with insulin (33.5% vs. 22.8%, P < 0.0001).
Table 1
Baseline characteristics of TECOS participants who did not have a within-trial nonfatal myocardial infarction (MI), and for those participants with a nonfatal MI, split by sitagliptin or placebo treatment
Characteristic
Patients without a nonfatal MI during the trial randomized to sitagliptin or placebo
N = 14,055
Patients with a nonfatal MI during the trial
P-value*
Sitagliptin
N = 289
Placebo
N = 302
Age at randomization (years)a
65.0 (60.0, 71.0)
67.0 (62.0, 74.0)
66.0 (60.0, 72.0)
0.1414
Female
4161 (29.6%)
56 (19.4%)
75 (24.8%)
0.1103
Hispanic or Latino
1754 (12.5%)
17 (5.9%)
22 (7.3%)
0.4924
Race
0.4603
 White
9472 (67.4%)
235 (81.3%)
234 (77.5%)
 
 Black
422 (3.0%)
10 (3.5%)
14 (4.6%)
 
 Asian
3184 (22.7%)
38 (13.1%)
42 (13.9%)
 
 Other
977 (7.0%)
6 (2.1%)
12 (4.0%)
 
Region
0.5023
 Latin America
1445 (10.3%)
10 (3.5%)
11 (3.6%)
 
 Asia Pacific and other
4377 (31.1%)
98 (33.9%)
84 (27.8%)
 
 Western Europe
1977 (14.1%)
48 (16.6%)
50 (16.6%)
 
 Eastern Europe
3822 (27.2%)
63 (21.8%)
68 (22.5%)
 
 North America
2434 (17.3%)
70 (24.2%)
89 (29.5%)
 
Durationb of type 2 diabetes (years)
10.0 (5.0, 16.0)
11.0 (6.0, 18.0)
11.0 (6.0, 16.0)
0.3233
Diabetes therapy at baseline (alone or in combination)
 Sulfonylurea
6394 (45.5%)
110 (38.1%)
125 (41.4%)
0.4085
 Metformin
11,501 (81.8%)
212 (73.4%)
235 (77.8%)
0.2069
 Thiazolidinedione
376 (2.7%)
9 (3.1%)
9 (3.0%)
0.9245
 Insulin
3208 (22.8%)
98 (33.9%)
98 (32.5%)
0.7063
Preexisting vascular disease
13,975 (99.4%)
288 (99.7%)
302 (100.0%)
0.4890
Coronary artery disease
10,312 (73.4%)
261 (90.3%)
269 (89.1%)
0.6208
Cerebrovascular disease
3445 (24.5%)
72 (24.9%)
65 (21.5%)
0.3289
Peripheral arterial disease
2348 (16.7%)
40 (13.8%)
42 (13.9%)
0.9814
Prior MI
5899 (42.0%)
171 (59.2%)
170 (56.3%)
0.4790
Prior congestive heart failure
2511 (17.9%)
62 (21.5%)
61 (20.2%)
0.7072
Previous atrial fibrillation/flutter
1086 (7.7%)
32 (11.1%)
45 (14.9%)
0.1670
NYHA classification
0.3241
 1
510 (20.3%)
16 (25.8%)
8 (13.1%)
 
 2
1256 (50.0%)
24 (38.7%)
30 (49.2%)
 
 3
339 (13.5%)
6 (9.7%)
10 (16.4%)
 
 4
11 (0.4%)
1 (1.6%)
1 (1.6%)
 
 Not available
395 (15.7%)
15 (24.2%)
12 (19.7%)
 
Qualifying HbA1c (mmol/mol)
55.2 (50.8, 60.7)
56.1 (51.0, 61.7)
55.2 (51.9, 59.6)
0.3239
Qualifying HbA1c (%)
7.2 (6.8, 7.7)
7.3 (6.8, 7.8)
7.2 (6.9, 7.6)
0.3239
eGFR (mL/min/1.73 m2)
73.0 (60.0, 88.0)
68.5 (55.0, 84.0)
69.0 (56.0, 88.0)
0.2918
Urine albumin creatinine ratio (g/mol creatinine)
10.6 (3.5, 35.0)
12.2 (5.3, 52.7)
13.8 (5.3, 43.9)
0.9026
Heart rate (bpm)
72.0 (65.0, 79.0)
70.0 (62.0, 78.0)
71.0 (62.0, 80.0)
0.0595
Body mass index (kg/m2)
29.5 (26.3, 33.2)
29.8 (26.6, 33.5)
30.4 (27.2, 34.3)
0.1480
Weight (kg)
83.0 (71.0, 96.0)
85.0 (75.0, 98.0)
88.5 (75.0, 100.0)
0.1517
Height (cm)
168.0 (160.0, 174.2)
169.4 (163.2, 175.3)
170.0 (162.6, 176.0)
0.5036
Cigarette smoking status
0.9049
 Current
1589 (11.3%)
44 (15.2%)
43 (14.2%)
 
 Former
5575 (39.7%)
129 (44.6%)
133 (44.0%)
 
 Never
6891 (49.0%)
116 (40.1%)
126 (41.7%)
 
Systolic blood pressure (mmHg)
133.0 (124.0, 145.0)
136.0 (124.0, 146.0)
135.0 (124.0, 148.0)
0.9803
Diastolic blood pressure (mmHg)
79.0 (70.0, 84.0)
77.0 (68.0, 82.0)
76.0 (68.0, 85.0)
0.3050
LDL-C
84.0 (65.0, 109.0)
81.0 (63.0, 99.6)
82.1 (65.6, 108.1)
0.4128
Medications taken at time of randomization
 Statins
11,213 (79.8%)
238 (82.4%)
248 (82.1%)
0.9408
 ACE inhibitors or angiotensin receptor blockers
11,040 (78.5%)
238 (82.4%)
255 (84.4%)
0.4959
 Diuretics
5727 (40.7%)
127 (43.9%)
150 (49.7%)
0.1633
 Calcium channel blockers
4730 (33.7%)
104 (36.0%)
118 (39.1%)
0.4386
 Beta blockers
8876 (63.2%)
210 (72.7%)
221 (73.2%)
0.8881
 Aspirin
11,027 (78.5%)
244 (84.4%)
235 (77.8%)
0.0403
Data shown are median (interquartile range) or N (%)
ACE angiotensin-converting enzyme, eGFR estimated glomerular filtration rate, HbA1c glycated hemoglobin, LDL-C low-density lipoprotein cholesterol, NYHA New York Heart Association, TECOS Trial Evaluating Cardiovascular Outcomes with Sitagliptin
*P-value is for placebo vs sitagliptin in patients with a nonfatal MI
aAge is missing among patients enrolled in Lithuania because the entire birth date including year was not available
bDuration = (year of randomization − year of diagnosis) + 1

Fatal and nonfatal MI

A total of 616 (4.2%) of the 14,671 TECOS participants had a within-trial fatal or nonfatal MI (300 [49%] randomized to sitagliptin and 316 [51%] to placebo), with no significant difference in the time to first event by randomized therapy (HR 0.95, 95% CI 0.81–1.11, P = 0.49) as reported previously [7]. Outcome information was missing for one participant for hHF and for two other participants for atrial fibrillation and stroke, limiting the number of participants who could be analyzed for these outcomes to 615 and 614, respectively. Twenty-five of these first MI events were fatal, 11 in the sitagliptin group and 14 in the placebo group, leaving 289 and 302 participants respectively with nonfatal MIs. Of the 591 participants who had a within-trial nonfatal MI, 87 (15%) died subsequently (66 [11%] classified as CV death), 57 (10%) experienced hHF, 109 (18%) had a second MI, 20 (3%) had a stroke, and 37 (6%) had incident atrial fibrillation.

CV events after nonfatal MI

The composite outcome of CV death or hHF following a nonfatal MI occurred in 58 of 289 sitagliptin group participants (20.1%; 13.9 events per 100 person-years) and in 50 of 302 placebo group participants (16.6%; 11.7 per 100 person-years), with no significant difference between groups (HR 1.21, 95% CI 0.83–1.77, P = 0.32; adjusted HR 1.23, 95% CI 0.83–1.82, P = 0.31) (Fig. 1a and Table 2). Similar results were seen for the individual outcomes of CV death, hHF, incident heart failure, recurrent MI, and all-cause death, and for the extended composite (CV death, hHF, incident heart failure, recurrent MI, stroke, or incident atrial fibrillation), with no significant differences also seen after adjustment for potential confounders (Table 2). Results were also similar when fatal MI was included in the cohort of interest (Additional file 1: Table S2, Figure S1).
Table 2
Cardiovascular outcomes occurring after a first within-trial non-fatal myocardial infarction in those randomized previously to sitagliptin or placebo treatment (intention-to-treat analysis)
 
Sitagliptin
n = 289
Placebo
n = 302
Unadjusted hazard ratio (95% CI)
P-value
Adjusted hazard ratio (95% CI)
P-value
No. (%)
Events per 100 patient-years
No. (%)
Events per 100 patient-years
Cardiovascular death or hospitalization for heart failure
58 (20.1)
13.9
50 (16.6)
11.7
1.21 (0.83–1.77)
0.32
1.23 (0.83–1.82)
0.31
Cardiovascular death
34 (11.8)
7.6
32 (10.6)
7.1
1.11 (0.68–1.81)
0.67
1.12 (0.67–1.86)
0.67
Hospitalization for heart failure
31 (10.7)
7.5
26 (8.6)
6.1
1.26 (0.75–2.12)
0.39
1.40 (0.80–2.42)
0.23
New onset heart failure
19 (6.6)
4.3
17 (5.6)
3.8
1.25 (0.64–2.44)
0.51
1.49 (0.72–3.09)
0.28
Cardiovascular death, hospital admission for heart failure, new heart failure, acute myocardial infarction, stroke or new-onset atrial fibrillation
108 (37.4)
33.0
100 (33.1)
28.4
1.16 (0.89–1.53)
0.27
1.21 (0.91–1.60)
0.20
Further acute myocardial infarction
54 (18.7)
7.4
55 (18.2)
7.1
1.01 (0.69–1.48)
0.95
0.99 (0.67–1.46)
0.95
All-cause death
50 (17.3)
11.0
37 (12.3)
8.1
1.40 (0.92–2.15)
0.12
1.41 (0.90–2.21)
0.13

On-treatment sensitivity analyses

At the time of the first nonfatal MI, 249 (42%) participants were taking a DPP-4i and 341 (58%) were not. There was no significant difference in the composite outcome of CV death or hHF for those treated or not treated with a DPP-4i (Fig. 2a and Table 3) for either unadjusted analyses (HR 0.91, 95% CI 0.62–1.34, P = 0.63) or adjusted analyses (HR 0.95, 95% CI 0.64–1.43, P = 0.82). All results were consistent with those for the intention-to-treat analyses, although CV deaths were numerically less in those treated with a DPP4i (HR 0.75). Results were also consistent when first fatal MI was included in the analysis (Additional file 1: Table S3, Figure S2).
Table 3
Cardiovascular outcomes occurring after a first within-trial nonfatal myocardial infarction in those pretreated or not pretreated with a dipeptidyl peptidase-4 inhibitor (DPP-4i) (on-treatment sensitivity analysis)
 
DPP-4i treated
n = 249
Not DPP-4i treated
n = 341
Unadjusted hazard ratio (95% CI)
P-value
Adjusted hazard ratio (95% CI)
P-value
No. (%)
Events per 100 patient-years
No. (%)
Events per 100 patient-years
Cardiovascular death or hospitalization for heart failure
45 (18.1)
11.9
62 (18.2)
13.3
0.91 (0.62–1.34)
0.63
0.95 (0.64–1.43)
0.82
Cardiovascular death
25 (10.0)
6.2
40 (11.7)
8.1
0.78 (0.47–1.29)
0.34
0.75 (0.44–1.26)
0.27
Hospitalization for heart failure
27 (10.8)
7.2
30 (8.8)
6.4
1.15 (0.68–1.94)
0.60
1.34 (0.77–2.33)
0.31
New onset heart failure
16 (6.4)
4.0
20 (5.9)
4.1
1.05 (0.54–2.05)
0.88
1.34 (0.64–2.79)
0.44
Cardiovascular death, hospital admission for heart failure, new heart failure, acute myocardial infarction, stroke or new-onset atrial fibrillation
87 (34.9)
28.8
120 (35.2)
31.8
0.92 (0.70–1.22)
0.56
0.95 (0.71–1.27)
0.72
Further acute myocardial infarction
46 (18.5)
7.2
63 (18.5)
7.3
0.97 (0.66–1.42)
0.89
0.99 (0.67–1.46)
0.95
All-cause death
37 (14.9)
8.9
49 (14.4)
9.9
0.94 (0.61–1.44)
0.77
0.91 (0.58–1.43)
0.68

Discussion

Although preclinical data provided theoretical support [1922], these post hoc TECOS analyses found no evidence that treatment with sitagliptin, compared with placebo, given prior to a first within-trial nonfatal MI had any impact on subsequent CV outcomes. Similar results were obtained when previous use of any DPP-4i was examined, and in sensitivity analyses that included fatal as well as nonfatal MIs.
Possible explanations for the discordance between human and animal observations include the following: (1) all TECOS participants had established ASCVD versus the lack of disease in experimental animals; (2) our study had only modest statistical power with just 123 composite outcome events analyzed; (3) experimentally induced MI is typically the consequence of total occlusion of a large coronary vessel, leading to a rather large area of myocardial necrosis, associated with adverse clinical consequences and significant mortality in the animal models—in contrast, spontaneous acute MI in humans is more variable in terms of the size of the relevant coronary vessel and the corresponding size of the subtended myocardium, whether complete occlusion of the coronary occurs, and marked variability in the timing from MI onset to clinical presentation, all of which translates into highly variable sizes of the area at risk, i.e. receiving blood supply from the infarct-related vessel, and of the necrotic area [25, 26]; (4) the doses of sitagliptin used in the animal studies are roughly twofold or more higher [1922]; and (5) not all TECOS participants may have been adherent with respect to their study medication, and the GLP-1 receptor agonism augmented by DPP-4is does not have the same CV consequences in humans that has been demonstrated in animal studies [1522]. Our results, however, are supported by negative results reported from a similar analysis of the Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial examining effects of liraglutide versus placebo pretreatment on CV events following MI occurring during the trial [30].
Controversy persists regarding the effects of DPP-4is on heart failure risk, originating from the observation of an increased risk of hHF with saxagliptin in the Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus-Thrombolysis in Myocardial Infarction 53 (SAVOR-TIMI) 53 trial [31] with a similar non-significant trend in the Examination of Cardiovascular Outcomes with Alogliptin versus Standard of Care (EXAMINE) trial with alogliptin [32], but no hHF signal observed with sitagliptin [27] or linagliptin [33]. On the other hand, results from observational studies have yielded counter-observations, reporting lower hHF risk associated with DPP-4i use compared with GLP-1 receptor agonists, with no significant difference in patients with a history of heart failure [34], and no difference in the risk of hHF when DPP-4i use was compared with sulfonylurea [35]. If DPP-4i treatment increases heart failure risk, the mechanism remains elusive. By echocardiographic criteria, a trend toward worsening diastolic ventricular function was slowed with sitagliptin treatment [36]. As a potential reason for a heterogeneity in effects between different DPP-4is, a suppression of renal sodium-hydrogen exchanger 3 activity with agents that are excreted in the urine (sitagliptin, alogliptin and linagliptin) has been proposed to protect from DPP-4i–induced heart failure [37]. In the present analysis, in accord with prior results of no heart failure effects of sitagliptin in the overall TECOS cohort, no association between sitagliptin and heart failure events was observed post-MI [7, 8, 27]. Thus, sitagliptin seems to be safe in patients during and after acute MI. Whether this applies to other DPP-4is needs to be studied in dedicated analyses from the respective CV outcomes trials [5, 6, 10]. Along these lines, a meta-analysis of other CV outcomes trials with DPP-4is (e.g. SAVOR TIMI-53 [5], EXAMINE [6], CArdiovascular safety and Renal Microvascular outcomE study with LINAgliptin [CARMELINA] [9, 10], and CARdiovascular Outcome Trial of LINAgliptin Versus Glimepiride in Type 2 Diabetes [CAROLINA] [38]) could provide further clarification.
Limitations of the present analyses include the non-randomized selection of the subset with MI for analysis [7, 8]. In addition, incomplete adherence to randomized treatment that could have occurred selectively post-MI could further confound comparative analyses. These analyses had limited power given the relatively few patients with MI with subsequent outcomes of interest. However, this data set is larger than most available with an ability to explore such associations.

Conclusions

In summary, these post hoc analyses of data from TECOS participants who had type 2 diabetes and ASCVD do not support the preclinically derived hypothesis that DPP-4i treatment prior to an MI can reduce the subsequent risk of CV death or hHF.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12933-019-0921-2.

Acknowledgements

The authors acknowledge John Schibler for his help in generating graphics and formatting tables of this study. The authors thank Peter Hoffmann for excellent editorial support. RRH is an NIHR Senior Investigator.

Previous presentation

Poster at the 2017 Annual Meeting of the European Association for the Study of Diabetes.
The trial was designed and overseen by a steering committee, and an independent data and safety monitoring committee performed regular safety surveillance. All patients provided written informed consent. Institutional review board approval was required at all participating institutions.
Not applicable.

Competing interests

MAN has been a member of advisory boards or consulted for AstraZeneca (moderate), Boehringer Ingelheim (moderate), Eli Lilly & Co. (significant), Fractyl (moderate), GlaxoSmithKline (moderate), Intarcia (moderate), Menarini/Berlin Chemie (moderate), Merck, Sharp & Dohme (significant), and NovoNordisk (significant). His institution has received grant support from AstraZeneca, Boehringer Ingelheim, Eli Lilly & Co., GlaxoSmithKline, Intarcia, Menarini/Berlin-Chemie, Merck, Sharp & Dohme, Novartis Pharma, and Novo Nordisk A/S. He has also served on the speakers’ bureau of AstraZeneca, Boehringer Ingelheim, Eli Lilly & Co., GlaxoSmithKline, Menarini/Berlin Chemie (all moderate), Merck, Sharp & Dohme, and Novo Nordisk A/S (both significant). DKM has provided clinical trial leadership for AstraZeneca, Sanofi Aventis, Janssen, Boehringer Ingelheim, Merck & Co, Pfizer, Lilly US, Novo Nordisk, Lexicon, Eisai, GlaxoSmithKline, and Esperion, and consultancy for AstraZeneca, Sanofi Aventis, Lilly US, Boehringer Ingelheim, Merck & Co, Novo Nordisk, Applied Therapeutics, Afimmune and Metavant. KSP has no disclosures. YL has received grants from Merck, Janssen Research & Development, AstraZeneca, GlaxoSmithKline, and Bayer HealthCare AG. TES reports personal fees from several companies (incl. Amgen, AstraZeneca, Merck, NovoNordisk, Pfizer, Orion, Bayer, Boehringer-Ingelheim) and owns a minor amount of stock in OrionPharma. AR has no disclosures. TD has no disclosures. EDP has received grants from Janssen, Merck, Sanofi, AstraZeneca, Genentech, and Amgen, and has consulting associations with Janssen, Bayer, Merck, and Sanofi. HDW reports research grants from GlaxoSmithKline, Sanofi-Aventis, Eli Lilly and Company, National Institute of Health, Merck Sharp & Dohme, George Institute, Omthera Pharmaceuticals, Pfizer New Zealand, Intarcia Therapeutics Inc., Elsai Inc., Daiichi-Sankyo, DalCor Pharmaceuticals; Advisory board/lecture fees from AstraZeneca, Acetelion, Sirte and he is a Steering Board member for Luitpold Pharmaceuticals Ltd and CSL Behring LLC. RS has no disclosures. RRH reports receiving grants from AstraZeneca during the conduct of the study and grants and personal fees from Bayer, Boehringer Ingelheim and Merck Sharp&Dohme Corp., a subsidiary of Merck & Co., Inc.; personal fees from Novartis, Amgen, and Servier; and financial support from Elcelyx, GlaxoSmithKline, Janssen, and Takeda outside the submitted work.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Drucker DJ. The role of gut hormones in glucose homeostasis. J Clin Invest. 2007;117:24–32.CrossRef Drucker DJ. The role of gut hormones in glucose homeostasis. J Clin Invest. 2007;117:24–32.CrossRef
2.
Zurück zum Zitat Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368:1696–705.CrossRef Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368:1696–705.CrossRef
8.
Zurück zum Zitat Green JB, Bethel MA, Paul SK, et al. Rationale, design, and organization of a randomized, controlled Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS) in patients with type 2 diabetes and established cardiovascular disease. Am Heart J. 2013;166(983–9):e7. Green JB, Bethel MA, Paul SK, et al. Rationale, design, and organization of a randomized, controlled Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS) in patients with type 2 diabetes and established cardiovascular disease. Am Heart J. 2013;166(983–9):e7.
9.
Zurück zum Zitat Rosenstock J, Perkovic V, Alexander JH, CARMELINA® Investigators, et al. Rationale, design, and baseline characteristics of the CArdiovascular safety and Renal Microvascular outcomE study with LINAgliptin (CARMELINA®): a randomized, double-blind, placebo-controlled clinical trial in patients with type 2 diabetes and high cardio-renal risk. Cardiovasc Diabetol. 2018;17:39. https://doi.org/10.1186/s12933-018-0682-3.CrossRefPubMedPubMedCentral Rosenstock J, Perkovic V, Alexander JH, CARMELINA® Investigators, et al. Rationale, design, and baseline characteristics of the CArdiovascular safety and Renal Microvascular outcomE study with LINAgliptin (CARMELINA®): a randomized, double-blind, placebo-controlled clinical trial in patients with type 2 diabetes and high cardio-renal risk. Cardiovasc Diabetol. 2018;17:39. https://​doi.​org/​10.​1186/​s12933-018-0682-3.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Rosenstock J, Perkovic V, Johansen OE, et al. Effect of linagliptin vs placebo on major cardiovascular events in adults with type 2 diabetes and high cardiovascular and renal risk: the CARMELINA randomized clinical trial. JAMA. 2019;321:69–79.CrossRef Rosenstock J, Perkovic V, Johansen OE, et al. Effect of linagliptin vs placebo on major cardiovascular events in adults with type 2 diabetes and high cardiovascular and renal risk: the CARMELINA randomized clinical trial. JAMA. 2019;321:69–79.CrossRef
11.
Zurück zum Zitat Drucker DJ. The cardiovascular biology of glucagon-like peptide-1. Cell Metab. 2016;24:15–30.CrossRef Drucker DJ. The cardiovascular biology of glucagon-like peptide-1. Cell Metab. 2016;24:15–30.CrossRef
13.
Zurück zum Zitat Pujadas G, Drucker DJ. Vascular biology of glucagon receptor superfamily peptides: complexity, controversy, and clinical relevance. Endocr Rev. 2016;37:554–83.CrossRef Pujadas G, Drucker DJ. Vascular biology of glucagon receptor superfamily peptides: complexity, controversy, and clinical relevance. Endocr Rev. 2016;37:554–83.CrossRef
14.
Zurück zum Zitat Katakami N, Mita T, Irie Y, et al. Effect of sitagliptin on tissue characteristics of the carotid wall in patients with type 2 diabetes: a post hoc sub-analysis of the sitagliptin preventive study of intima-media thickness evaluation (SPIKE). Cardiovasc Diabetol. 2018;17:24.CrossRef Katakami N, Mita T, Irie Y, et al. Effect of sitagliptin on tissue characteristics of the carotid wall in patients with type 2 diabetes: a post hoc sub-analysis of the sitagliptin preventive study of intima-media thickness evaluation (SPIKE). Cardiovasc Diabetol. 2018;17:24.CrossRef
15.
Zurück zum Zitat Bose AK, Mocanu MM, Carr RD, Brand CL, Yellon DM. Glucagon-like peptide 1 can directly protect the heart against ischemia/reperfusion injury. Diabetes. 2005;54:146–51.CrossRef Bose AK, Mocanu MM, Carr RD, Brand CL, Yellon DM. Glucagon-like peptide 1 can directly protect the heart against ischemia/reperfusion injury. Diabetes. 2005;54:146–51.CrossRef
16.
Zurück zum Zitat Bose AK, Mocanu MM, Carr RD, Yellon DM. Glucagon like peptide-1 is protective against myocardial ischemia/reperfusion injury when given either as a preconditioning mimetic or at reperfusion in an isolated rat heart model. Cardiovasc Drugs Ther. 2005;19:9–11.CrossRef Bose AK, Mocanu MM, Carr RD, Yellon DM. Glucagon like peptide-1 is protective against myocardial ischemia/reperfusion injury when given either as a preconditioning mimetic or at reperfusion in an isolated rat heart model. Cardiovasc Drugs Ther. 2005;19:9–11.CrossRef
17.
Zurück zum Zitat Noyan-Ashraf MH, Momen MA, Ban K, et al. GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes. 2009;58:975–83.CrossRef Noyan-Ashraf MH, Momen MA, Ban K, et al. GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes. 2009;58:975–83.CrossRef
20.
Zurück zum Zitat Chinda K, Sanit J, Chattipakorn S, Chattipakorn N. Dipeptidyl peptidase-4 inhibitor reduces infarct size and preserves cardiac function via mitochondrial protection in ischaemia-reperfusion rat heart. Diabetes Vasc Dis Res. 2014;11:75–83.CrossRef Chinda K, Sanit J, Chattipakorn S, Chattipakorn N. Dipeptidyl peptidase-4 inhibitor reduces infarct size and preserves cardiac function via mitochondrial protection in ischaemia-reperfusion rat heart. Diabetes Vasc Dis Res. 2014;11:75–83.CrossRef
21.
Zurück zum Zitat Chinda K, Palee S, Surinkaew S, Phornphutkul M, Chattipakorn S, Chattipakorn N. Cardioprotective effect of dipeptidyl peptidase-4 inhibitor during ischemia-reperfusion injury. Int J Cardiol. 2013;167:451–7.CrossRef Chinda K, Palee S, Surinkaew S, Phornphutkul M, Chattipakorn S, Chattipakorn N. Cardioprotective effect of dipeptidyl peptidase-4 inhibitor during ischemia-reperfusion injury. Int J Cardiol. 2013;167:451–7.CrossRef
23.
Zurück zum Zitat McCormick LM, Kydd AC, Read PA, et al. Chronic dipeptidyl peptidase-4 inhibition with sitagliptin is associated with sustained protection against ischemic left ventricular dysfunction in a pilot study of patients with type 2 diabetes mellitus and coronary artery disease. Circ Cardiovasc Imaging. 2014;7:274–81.CrossRef McCormick LM, Kydd AC, Read PA, et al. Chronic dipeptidyl peptidase-4 inhibition with sitagliptin is associated with sustained protection against ischemic left ventricular dysfunction in a pilot study of patients with type 2 diabetes mellitus and coronary artery disease. Circ Cardiovasc Imaging. 2014;7:274–81.CrossRef
24.
Zurück zum Zitat Read PA, Khan FZ, Heck PM, Hoole SP, Dutka DP. DPP-4 inhibition by sitagliptin improves the myocardial response to dobutamine stress and mitigates stunning in a pilot study of patients with coronary artery disease. Circ Cardiovasc Imaging. 2010;3:195–201.CrossRef Read PA, Khan FZ, Heck PM, Hoole SP, Dutka DP. DPP-4 inhibition by sitagliptin improves the myocardial response to dobutamine stress and mitigates stunning in a pilot study of patients with coronary artery disease. Circ Cardiovasc Imaging. 2010;3:195–201.CrossRef
25.
Zurück zum Zitat Hartman MHT, Eppinga RN, Vlaar PJJ, et al. The contemporary value of peak creatine kinase-MB after ST-segment elevation myocardial infarction above other clinical and angiographic characteristics in predicting infarct size, left ventricular ejection fraction, and mortality. Clin Cardiol. 2017;40:322–8. https://doi.org/10.1002/clc.22663.CrossRefPubMed Hartman MHT, Eppinga RN, Vlaar PJJ, et al. The contemporary value of peak creatine kinase-MB after ST-segment elevation myocardial infarction above other clinical and angiographic characteristics in predicting infarct size, left ventricular ejection fraction, and mortality. Clin Cardiol. 2017;40:322–8. https://​doi.​org/​10.​1002/​clc.​22663.CrossRefPubMed
27.
Zurück zum Zitat McGuire DK, Van de Werf F, Armstrong PW, Trial Evaluating Cardiovascular Outcomes With Sitagliptin (TECOS) Study Group, et al. Association between sitagliptin use and heart failure hospitalization and related outcomes in type 2 diabetes mellitus: secondary analysis of a randomized clinical trial. JAMA Cardiol. 2016;1:126–35. https://doi.org/10.1001/jamacardio.2016.0103.CrossRefPubMed McGuire DK, Van de Werf F, Armstrong PW, Trial Evaluating Cardiovascular Outcomes With Sitagliptin (TECOS) Study Group, et al. Association between sitagliptin use and heart failure hospitalization and related outcomes in type 2 diabetes mellitus: secondary analysis of a randomized clinical trial. JAMA Cardiol. 2016;1:126–35. https://​doi.​org/​10.​1001/​jamacardio.​2016.​0103.CrossRefPubMed
30.
Zurück zum Zitat Nauck MA, Tornøe K, Rasmussen S, Bach Treppendahl M, Marso SP, LEADER Publication Committee on behalf of the LEADER Trial Investigators. Cardiovascular outcomes in patients who experienced a myocardial infarction while treated with liraglutide versus placebo in the LEADER trial. Diabetes Vasc Dis Res. 2018;15:465–8. https://doi.org/10.1177/1479164118783935.CrossRef Nauck MA, Tornøe K, Rasmussen S, Bach Treppendahl M, Marso SP, LEADER Publication Committee on behalf of the LEADER Trial Investigators. Cardiovascular outcomes in patients who experienced a myocardial infarction while treated with liraglutide versus placebo in the LEADER trial. Diabetes Vasc Dis Res. 2018;15:465–8. https://​doi.​org/​10.​1177/​1479164118783935​.CrossRef
31.
Zurück zum Zitat Scirica BM, Braunwald E, Raz I, et al. Heart failure, saxagliptin, and diabetes mellitus: observations from the SAVOR-TIMI 53 randomized trial. Circulation. 2014;130:1579–88.CrossRef Scirica BM, Braunwald E, Raz I, et al. Heart failure, saxagliptin, and diabetes mellitus: observations from the SAVOR-TIMI 53 randomized trial. Circulation. 2014;130:1579–88.CrossRef
32.
Zurück zum Zitat Zannad F, Cannon CP, Cushman WC, et al. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet. 2015;385:2067–76.CrossRef Zannad F, Cannon CP, Cushman WC, et al. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet. 2015;385:2067–76.CrossRef
33.
Zurück zum Zitat McGuire DK, Alexander JH, Johansen OE, et al. Linagliptin effects on heart failure and related outcomes in individuals with type 2 diabetes mellitus at high cardiovascular and renal risk in CARMELINA. Circulation. 2019;139:351–61.CrossRef McGuire DK, Alexander JH, Johansen OE, et al. Linagliptin effects on heart failure and related outcomes in individuals with type 2 diabetes mellitus at high cardiovascular and renal risk in CARMELINA. Circulation. 2019;139:351–61.CrossRef
34.
Zurück zum Zitat Dawwas GK, Smith SM, Park H. Risk of heart failure hospitalization among users of dipeptidyl peptidase-4 inhibitors compared to glucagon-like peptide-1 receptor agonists. Cardiovasc Diabetol. 2018;17:102.CrossRef Dawwas GK, Smith SM, Park H. Risk of heart failure hospitalization among users of dipeptidyl peptidase-4 inhibitors compared to glucagon-like peptide-1 receptor agonists. Cardiovasc Diabetol. 2018;17:102.CrossRef
35.
Zurück zum Zitat Kim YG, Yoon D, Park S, et al. Dipeptidyl peptidase-4 inhibitors and risk of heart failure in patients with type 2 diabetes mellitus: A population-based cohort study. Circ Heart Fail. 2017;10:e003957.CrossRef Kim YG, Yoon D, Park S, et al. Dipeptidyl peptidase-4 inhibitors and risk of heart failure in patients with type 2 diabetes mellitus: A population-based cohort study. Circ Heart Fail. 2017;10:e003957.CrossRef
36.
Zurück zum Zitat Yamada H, Tanaka A, Kusunose K, et al. Effect of sitagliptin on the echocardiographic parameters of left ventricular diastolic function in patients with type 2 diabetes: a subgroup analysis of the PROLOGUE study. Cardiovasc Diabetol. 2017;16:63.CrossRef Yamada H, Tanaka A, Kusunose K, et al. Effect of sitagliptin on the echocardiographic parameters of left ventricular diastolic function in patients with type 2 diabetes: a subgroup analysis of the PROLOGUE study. Cardiovasc Diabetol. 2017;16:63.CrossRef
37.
Zurück zum Zitat Sano M. Mechanism by which dipeptidyl peptidase-4 inhibitors increase the risk of heart failure and possible differences in heart failure risk. J Cardiol. 2019;73:28–32.CrossRef Sano M. Mechanism by which dipeptidyl peptidase-4 inhibitors increase the risk of heart failure and possible differences in heart failure risk. J Cardiol. 2019;73:28–32.CrossRef
Metadaten
Titel
Sitagliptin does not reduce the risk of cardiovascular death or hospitalization for heart failure following myocardial infarction in patients with diabetes: observations from TECOS
verfasst von
Michael A. Nauck
Darren K. McGuire
Karen S. Pieper
Yuliya Lokhnygina
Timo E. Strandberg
Axel Riefflin
Tuncay Delibasi
Eric D. Peterson
Harvey D. White
Russell Scott
Rury R. Holman
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Cardiovascular Diabetology / Ausgabe 1/2019
Elektronische ISSN: 1475-2840
DOI
https://doi.org/10.1186/s12933-019-0921-2

Weitere Artikel der Ausgabe 1/2019

Cardiovascular Diabetology 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.