Skip to main content
Erschienen in: BMC Cardiovascular Disorders 1/2021

Open Access 01.12.2021 | Research article

Intermittent concurrent use of clopidogrel and proton pump inhibitors did not increase risk of adverse clinical outcomes in Chinese patients with coronary artery disease

verfasst von: Wanbing He, Xiaorong Shu, Enyi Zhu, Bingqing Deng, Yongqing Lin, Xiaoying Wu, Zenan Zhou, Jingfeng Wang, Ruqiong Nie

Erschienen in: BMC Cardiovascular Disorders | Ausgabe 1/2021

Abstract

Background

Proton pump inhibitors (PPIs) are frequently prescribed to patients with coronary heart disease (CHD) under antiplatelet therapy to prevent gastrointestinal (GI) bleeding. However, its clinical impact is still under debate, especially in Asian population. This study was undertaken to explore the effects of concurrent use of clopidogrel and PPIs on the clinical outcomes in Chinese patients with CHD in secondary prevention.

Methods

A single-center retrospective study was conducted in 638 patients with CHD on consecutive clopidogrel therapy for at least 1 year. After 18-month follow-up, adverse clinical events were collected. Cox regression was used to calculate hazard ratios (HR) and 95% confidence interval (CI) for the effect of PPI use on the outcomes. A total of 638 patients were recruited from 2014 to 2015 in this study, among whom 201 were sustained PPI users, 188 were intermittent PPI users and the remaining 249 were non-PPI users.

Results

Compared with sustained PPI users, intermittent use of PPIs was associated with a lower risk of stroke, major adverse cardiac events (MACE) and net adverse clinical event (NACE) (stroke: adjusted HR: 0.109, 95% CI 0.014–0.878, p = 0.037; MACE: adjusted HR: 0.293, 95% CI 0.119–0.722; p = 0.008; NACE: adjusted HR: 0.357, 95% CI 0.162–0.786, p = 0.011). Subgroup analysis further revealed the benefit of intermittent PPI use was significant in male CHD patients over 60 years old, with hypertension or chronic kidney disease, and undergoing percutaneous coronary intervention during hospitalization.

Conclusion

The current findings suggest that the intermittent concurrent use of PPIs and clopidogrel is not associated with an increased risk of 18-month adverse clinical outcomes, and intermittent use of PPIs is associated with a lower rate of MACE and NACE.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12872-021-01884-z.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACEI
Angiotensin converting enzyme inhibitor
ACS
Acute coronary syndrome
AF
Atrial fibrillation
ALT
Alanine transaminase
AMI
Acute myocardial infarction
ANOVA
Analysis of variance
ARB
Angiotensin II receptor antagonist
AST
Glutamic-oxaloacetic transaminase
BARC
Bleeding Academic Research Consortium
BMI
Body mass index
BP
Blood pressure
CCB
Calcium channel blocker
CHD
Coronary heart disease
CHF
Chronic heart failure
CI
Confidence interval
CKD
Chronic kidney disease
CK-MB
Creatine phosphokinase isoenzyme
CVD
Cardiovascular diseases
CYP
Cytochrome
DAPT
Dual antiplatelet therapy
DM
Diabetes mellitus
EMs
Extensive (normal) metabolizers
FPG
Fasting plasma glucose
GERD
Gastroesophageal reflux disease
GI
Gastrointestinal
GOF
Gain-of-function
HDL-C
High-density lipoprotein cholesterol
HR
Hazard ratios
hs-CRP
High-sensitivity C-reactive protein
IMs
Intermediate metabolizers
LDL-C
Low-density lipoprotein cholesterol
MACE
Major adverse cardiac event
MI
Myocardial infarction
NACE
Net adverse clinical event
NSAIDS
Non-steroidal anti-inflammatory drugs
PCI
Percutaneous coronary intervention
PMs
Poor metabolizers
PPIs
Proton pump inhibitors
RCTs
Randomized controlled trials
SD
Standard deviation
TG
Triglyceride
TIA
Transient ischemic attack
TC
Total cholesterol
UMs
Ultra-rapid metabolizers

Background

Cardiovascular diseases (CVD) have become the leading causes of death worldwide. According to the 2013 Global Burden of Disease Study, approximately 17 million people died of CVD worldwide, accounting for approximately 31.5% of the total number of deaths [1]. Among the different types of CVDs, the mortality of coronary artery disease (CHD) was the highest [2, 3]. Therefore, it is important to prevent CHD.
The rupture or erosion of vulnerable plaques that induces platelet activation and aggregation are the key pathophysiological mechanisms, which contributes to the deaths of patients with CHD. Therefore, antiplatelet therapy is critical to reduce cardiovascular deaths in patients with CHD [4]. Clopidogrel combined with aspirin, called dual antiplatelet therapy (DAPT), is the basic treatment strategy for CHD, especially for acute coronary syndrome (ACS). DAPT can effectively prevent percutaneous coronary intervention (PCI) from in-stent thrombosis and reduce cardiovascular adverse events. Nevertheless, the risks of gastrointestinal (GI) and other bleeding events are significantly increased after treatment with DAPT. Thus, patients who accept DAPT, especially after PCI, often take proton pump inhibitors (PPIs) at the same time. Most PPIs such as omeprazole, esomeprazole and lansoprazole, are mainly metabolized by liver cytochrome (CYP) P450 isoenzyme CYP2C19, which also regulates clopidogrel metabolism. When combined with clopidogrel, PPIs may exert a competitive inhibitory effect, and thus affect the therapeutic effects of clopidogrel. Previous pharmacodynamic studies showed that the concurrent use of PPIs could reduce anti-platelet activities of clopidogrel [58]. However, the results from clinical trials were inconsistent. Some large clinical studies such as ADAPT-DES, BASKET, and CAPRIE indicated that combined use was associated with an increased risk of cardiovascular adverse events [4, 9, 10]. However, the results from other large-scale prospective clinical studies such as COGENT, CREDO, PRINCIPLE-TIMI 44, PRODIGY, TRITON-TIMI 38 showed the opposite results, which indicated that the concurrent use of PPIs and clopidogrel did not increase the risk of net adverse clinical events (NACE) and even had benefits by reducing the risk of GI bleeding [7, 9, 11, 12]. Furthermore, clopidogrel was more frequently used in combination with PPIs in Asian countries than that in Western countries [13, 14]. However, researches on the clinical effect of the concurrent use on both cardiovascular adverse events and GI bleeding in the Asian population are very limited. Therefore, this study was aimed to investigate the effect of concurrent use of clopidogrel and PPIs on clinical outcomes (composite endpoint events such as cardiovascular events and GI bleeding) in Chinese patients with CHD.

Materials and methods

Study population

This was a single-center retrospective study of consecutive CHD patients referred to the Sun Yat-sen Memorial Hospital of Sun Yat-sen University between January 2014 and April 2015. Patients over 18 years old with diagnosed CHD and under clopidogrel (75 mg/d) treatment over 12 months were included. Patients who met one of the following criteria were excluded: (1) anemia (hemoglobin < 90 g/L); (2) platelet count < 100 × 109/L, or with hematological diseases, which affect the number or function of platelets; (3) severe liver insufficiency with alanine transaminase (ALT) or glutamic-oxaloacetic transaminase (AST) three times higher than normal level; (4) under long-term non-steroidal anti-inflammatory drugs (NSAIDS) (except aspirin) or glucocorticoid treatment; (5) transferred to other hospital for treatment other than death, or the information for drug use can not be collected.
The follow-up study was conducted at 12 and 18 months via outpatient follow-up or telephone follow-up. The follow-up content included all-cause deaths, cardiovascular events, gastrointestinal side effects, gastrointestinal bleeding, chest pain and other discomfort symptoms. The consumption of drugs (clopidogrel and PPIs) during the follow-up was obtained via the outpatient prescription system. In addition, the reasons for those patients being not in the hospital for continuous hospitalization were also collected via telephone follow-up.
The study protocol conformed to the ethical guidelines of the 1975 Declaration of Helsinki as reflected in a priori approval by the Ethics Committee of Sun Yat-sen Memorial Hospital of Sun Yat-sen University. Informed consent was obtained from each participant.

Baseline clinical data

In this study, the hospitalization information was collected by querying the medical record system of the hospital. If the patient had multiple hospitalization records, only the most relevant and the earliest record within the enrolling period were chosen as baseline data. The following data were collected: (1) Demographic data: age, sex, body mass index (BMI), blood pressure (BP), smoking and alcohol status; (2) Comorbidity: CHD, acute myocardial infarction (AMI), chronic heart failure (CHF), hypertension, hyperlipidemia, atrial fibrillation (AF), transient ischemic attack (TIA), stroke, diabetes mellitus (DM), chronic kidney disease (CKD), gastroduodenal ulcer, gastrointestinal hemorrhage et al. (3) Surgical history: PCI, coronary artery bypass graft (CABG), cardiac device implantation, or other surgical history.

Laboratory parameters

Laboratory parameters were all measured by using blood sample. Each patient were fasted overnight for at least 10 h before venipuncture. Platelets counts, serum creatinine, fasting plasma glucose (FPG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), triglyceride (TG), high-sensitivity C-reactive protein (hs-CRP) and creatine phosphokinase isoenzyme (CK-MB) were analyzed by a standardized and certified TBA-120 auto-analyzer (Toshiba Medical Systems, Japan) in the institutional central laboratory. The CYP2C19 genotype was also detected. According to the current international classification, different CYP2C19 genotypes can be classified into four metabolic types: (1) extensive (normal) metabolizers (EMs), carrying two normal alleles (such as *1/*1); (2) intermediate metabolizers (IMs), carrying one loss-of-function (LOF) allele (e.g. *1/*2); (3) poor metabolizers (PMs), carrying two LOF alleles (e.g. *2/*2, *2/*3, *3/*3); (4) ultra-rapid metabolizers (UMs), carrying one or two gain-of-function (GOF) allele (such as *1/*17, *17/*17) [15]. The frequency of GOF allele in Chinese population is very low (about 1/1000), and thus this type of genotype was not detected in this study [16].

Drug use

The PPIs regimens were determined by the clinicians according to the actual condition of the patient and were not affected by this study. The dosage and frequency of PPIs were recorded in each patient’s visit. According to the length of time of taking PPIs, this study divided the patients into three groups: (1) Sustained PPI users: use of PPIs during the follow-up period more than 30 days; (2) Intermittent PPI users: ever use of PPIs during follow-up period but less than 30 days; (3) non-PPI users: no PPIs were taken during the follow-up period. Use of other drugs such as aspirin, angiotensin converting enzyme inhibitor (ACEI), angiotensin II receptor antagonist (ARB), statins, β-blockers, calcium channel blocker (CCB) were also collected according to the records of discharge from hospital. All included patients were under a standardized treatment of CHD secondary prevention determined by the clinicians blinded to this study.

Clinical outcomes

Clinical outcomes included all-cause death, re-hospitalization, myocardial infarction (MI), target vessel revascularization, stroke (ischemic), GI events or other bleeding events. GI events include: (1) symptoms of GI upset and diagnosis of GI diseases including gastritis or duodenitis, gastric or duodenal ulcer, gastric or duodenal obstruction, gastric or duodenal perforation; (2) GI bleeding: symptoms of hematemesis, melena, positive fecal occult blood, or the appearance of active bleeding under gastroscopy; (3) gastroesophageal reflux disease (GERD) determined by gastroscopy. Other bleeding events were defined as types 2, 3, and 5 in the 2011 Bleeding Academic Research Consortium (BARC) bleeding standard [17]. Major adverse cardiac event (MACE) was defined as the sum of cardiac deaths, MI, target vessel revascularization and stroke. Net adverse clinical event (NACE) was defined as the sum of all-cause deaths, MI, target vessel revascularization, stroke, GI bleeding, and BARC type 2, 3, and 5 events.

Statistical analysis

Data were presented as frequencies for categorical variables, mean values with standard deviation (SD) for normally distributed continuous variables and median values with 25th and 75th percentiles for ordinal variables. The comparison of continuous variables among multiple groups was performed by one-way analysis of variance (ANOVA) or Kruskal–Wallis test; while Chi-square test was performed for the comparison of categorical variables. The univariate Kaplan–Meier method was used to analyze the survival free from the clinical events, and the overall survival rate was compared using log-rank test. Cox proportional hazard regression model was used to adjust all possible confounding factors, which might influence the outcome of clinical events. In this study, the confounding factors included in the model were based on the differences among three groups and the factors associated with the occurrence of clinical events. The final factors included in the adjusted model were BMI, history of MI, stroke, DM, gastroduodenal ulcer, and alcohol consumption, whether to take β-blockers, aspirin, hemoglobin, whether to be hospitalized PCI. The final results were expressed as adjusted hazard ratio (adjusted HR) and its 95% confidence interval (95% CI). In order to further analyze the factors that affect the occurrence of clinical events, sensitivity analysis on the occurrence of NACE was also conducted. Subgroups were defined as age (≤ 6 0 years old and > 60 years old), gender (male, female), and hypertension, stroke, DM, CKD, gastroduodenal ulcer, type of PPIs (pantoprazole and other types), hospitalization for PCI, and CYP2C19 genotypes (EM, IM, PM) and compared by using the adjusted Cox proportional hazard regression model mentioned above. All statistical analyses were performed using SPSS 22.0 statistical software (SPSS, Inc, Chicago, IL). p < 0.05 was considered as statistical significance.

Results

Baseline characteristics of patients included

A total of 1151 patients with CHD were included. After excluding 513 patients who did not meet the inclusion criteria, the final study cohort consisted of 638 patients (Fig. 1). According to the duration of PPIs treatment, we divided the patients into three groups: 201 were sustained PPI users, 188 were intermittent PPI users, and the remaining 249 were non-PPI users. Tables 1, 2 and 3 summarized the baseline characteristics, medical history and laboratory parameters of enrolled patients among three groups, respectively. Patients with sustained PPI use suffered with the highest rate of MI but the lowest level of BMI, the least rate of DM; while those without PPI use exhibited the highest rate of stroke (Table 1). Patients with sustained PPI use suffered more gastroduodenal ulcer than the other two groups (Table 1). Particularly, although sustained PPI users tended to have higher rate of aspirin treatment than the other two, no significant differences were found among these three groups (Table 2). Among the patients with PPIs treatment, about 50.0% used pantoprazole, 12.6% used lansoprazole, and about 20.1% patients used two or more types of PPIs during the follow-up period (Table 2). The percentages of lansoprazole, omeprazole and esomeprazole treatment were significantly different between sustained and intermittent PPI users (Table 2; Fig. 2). Moreover, sustained PPI users tended to use two or more types of PPIs than the intermittent ones (Fig. 2). There were no differences among three groups in other variables such as age, sex, blood pressure, smoking state (Table 1), CYP2C19 genotypes or the laboratory indexes (Table 3).
Table 1
Baseline characteristics of patients with sustained, intermittent and non-PPI users
 
Sustained PPI users
Intermittent PPI users
Non-PPI users
p value
N
201
188
249
 
Demographic data
    
 Age (y)
66.9 ± 11.1
64.4 ± 11.4
65.2 ± 10.7
0.080
 Male (n, %)
123 (61.2%)
116 (61.7%)
174 (69.9%)
0.093
 BMI (kg/m2)
23.75 ± 4.30
24.17 ± 3.86
24.76 ± 2.97
0.022
 SBP (mmHg)
132.9 ± 20.8
134.8 ± 23.3
136.7 ± 21.1
0.180
 DBP (mmHg)
75.4 ± 12.4
77.1 ± 13.3
76.5 ± 12.3
0.408
Smoking
   
0.880
 Current
58 (28.9%)
60 (31.9%)
71 (28.5%)
 
 Former
31 (15.4%)
24 (12.8%)
39 (15.7%)
 
 None
112 (55.7%)
104 (55.3%)
139 (55.8%)
 
Alcohol
   
0.037
 Current
16 (8.0%)
15 (8.0%)
7 (2.8%)
 
 Former
5 (2.5%)
10 (5.3%)
15 (6.0%)
 
 None
180 (89.6%)
163 (86.7%)
227 (91.2%)
 
Comorbidity
    
 CHD
82 (40.8%)
57 (30.3%)
94 (37.8%)
0.088
 MI
35 (17.4%)
16 (8.5%)
35 (14.1%)
0.035
 CHF
10 (5.0%)
10 (5.3%)
11 (4.4%)
0.906
 CHD family history
26 (12.9%)
15 (8.0%)
22 (8.8%)
0.204
 Hypertension
144 (71.6%)
128 (68.1%)
179 (71.9%)
0.645
 Hypercholesterolemia
39 (19.4%)
34 (18.1%)
53 (21.3%)
0.700
 Atrial fibrosis
15 (7.5%)
11 (5.9%)
7 (2.8%)
0.076
 TIA
1 (0.5%)
2 (1.1%)
1 (0.4%)
0.659
 Stroke
34 (16.9%)
19 (10.1%)
47 (18.9%)
0.037
 DM
43 (21.4%)
61 (32.4%)
80 (32.1%)
0.019
 CKD
12 (6.0%)
17 (9.0%)
16 (6.4%)
0.440
 Gastroduodenal ulcer
45 (22.4%)
28 (14.9%)
20 (8.0%)
 < 0.001
 GI bleeding
4 (2.0%)
2 (1.1%)
4 (1.6%)
0.762
Surgical history
    
 PCI
50 (24.9%)
39 (20.7%)
70 (28.1%)
0.211
 CABG
0 (0.0%)
1 (0.5%)
2 (0.8%)
0.460
 Device implantation
6 (3.0%)
0 (0.0%)
7 (2.8%)
0.062
 Other surgical history
32 (15.9%)
30 (16.0%)
45 (18.1%)
0.781
BMI, body mass index; CABG, coronary artery bypass graft; CHD, coronary heart disease; CHF, chronic heart failure; CKD, chronic kidney disease; DBP, diastolic blood pressure; DM, diabetes mellitus; GI, gastrointestinal; MI, myocardial infarction; PCI, percutaneous coronary intervention; PPI, proton pump inhibitor; SBP, systolic blood pressure; TIA, transient ischemic attack
Table 2
Medical history of patients with sustained, intermittent and non-PPI users
 
Sustained PPI users
Intermittent PPI users
Non-PPI users
p value
N
201
188
249
 
Medicine history
    
 Aspirin
149 (74.1%)
125 (66.5%)
170 (68.3%)
0.221
PPI
    
 Pantoprazole
97 (48.2%)
96 (51.1%)
0.228
 Rabeprazole
9 (4.5%)
14 (7.4%)
0.399
 Lansoprazole
31 (15.4%)
18 (9.6%)
0.017
 Omeprazole
7 (3.5%)
18 (9.6%)
0.041
 Esomeprazole
4 (2.0%)
17 (9.0%)
0.007
 Two or more types
53 (26.4%)
25 (13.3%)
 < 0.001
ACEI/ARB
140 (69.7%)
120 (63.8%)
152 (61.0%)
0.160
CCB
98 (48.8%)
78 (41.5%)
112 (45.0%)
0.354
β blocker
162 (80.6%)
133 (70.7%)
174 (69.9%)
0.022
Statin
192 (95.5%)
174 (92.6%)
223 (89.6%)
0.061
Warfarin
4 (2.0%)
1 (0.5%)
1 (0.4%)
0.175
ACEI, angiotensin converting enzyme inhibitor; ARB, angiotensin II receptor antagonist; CCB, calcium channel blocker; PPI, proton pump inhibitor
Table 3
Laboratory parameters of patients with sustained, intermittent and non-PPI users
 
Sustained PPI users
Intermittent PPI users
Non-PPI users
p value
N
201
188
249
 
Laboratory parameters
    
Creatinine (mg/dL)
104.5 ± 70.8
111.3 ± 97.8
103.2 ± 56.7
0.552
Glucose (mmol/L)
5.5 ± 1.8
5.9 ± 2.4
5.7 ± 1.9
0.171
Total cholesterol (mmol/L)
4.22 (3.51, 5.11)
4.48 (3.81, 5.42)
4.47 (3.65, 5.25)
0.597
HDL-C (mmol/L)
1.07 (0.90, 1.32)
1.08 (0.89, 1.29)
1.06 (0.90, 1.26)
0.585
LDL-C (mmol/L)
2.46 (1.98, 3.12)
2.64 (2.14, 3.29)
2.59 (2.08, 3.19)
0.723
Triglycerides (mmol/L)
1.20 (0.94, 1.86)
1.38 (0.94, 2.04)
1.45 (1.05, 2.09)
0.239
Uric acid (µmol/L)
368.0 (311.0, 446.0)
393.0 (319.0, 477.0)
391.0 (329.0, 469.5)
0.916
ALT (U/L)
21.0 (15.0, 33.0)
20.0 (14.0, 35.5)
21.0 (15.0, 32.0)
0.455
AST (U/T)
23.0 (18.0, 38.0)
21.0 (18.0, 34.5)
23.0 (18.0, 29.0)
0.954
CK (U/L)
94.0 (64.0, 162.0)
87.0 (62.5, 136.0)
90.5 (65.0, 168.5)
0.758
CK-MB (U/L)
14.0 (11.0, 20.0)
14.0 (11.0, 19.0)
13.0 (11.0, 17.0)
0.567
hs-CRP (mg/L)
7156.0 (5888.0, 8098.0)
7439.0 (6282.0, 8259.0)
7544.5 (6269.8, 8507.0)
0.228
HbAlc (%)
6.0 (5.6, 6.6)
6.0 (5.6, 7.1)
6.1 (5.6, 7.1)
0.580
NT-proBNP
126.3 (55.1, 674.7)
151.0 (66.4, 587.6)
159.9 (63.2, 697.5)
0.732
TnT-HS
7.4 (4.4, 50.7)
13.2 (5.0, 398.0)
11.1 (5.4, 67.3)
0.439
Hb (g/L)
131.5 ± 19.1
129.4 ± 18.7
134.7 ± 22.5
0.042
Platelet count (× 109/L)
221.6 ± 60.8
235.5 ± 64.3
221.9 ± 62.2
0.059
CYP2C19 genotypes (%)
110
111
115
0.536
 EM
31.8
39.6
42.6
 
 IM
50.0
45.0
40.9
 
 PM
18.2
15.3
16.5
 
ALT, alanine transaminase; AST, aspartate aminotransferase; CK, creatine kinase; CK-MB, creatine kinase isoenzymes; EM, extensive metabolizers; Hb, hemoglobin; HbAlc, glycated hemoglobin; HDL-C, high-density lipoprotein cholesterol; hs-CRP, hyper-sensitive C-reactive protein; IM, intermediate metabolizers; LDL-C, low-density lipoprotein cholesterol; NT-proBNP, N-terminal (NT)-pro hormone BNP; PM, poor metabolizers; TNT-HS, high sensitive troponin T

Effect of PPIs use on clinical outcomes after 18-month follow-up

As shown in Table 4, patients with sustained PPIs use had the highest rate of 18-month stroke and re-hospitalization, mainly complaint of angina (Table 4). Stroke incidence was also the highest in sustained PPI users (Table 4). The Kaplan–Meier analysis revealed that PPIs use was significantly associated with 18-month rate of stoke (log rank p = 0.036; Fig. 3d). Further multivariable Cox regression analysis confirmed that intermittent use of PPIs was associated with lower risk of stroke compared with the sustained ones (adjusted HR: 0.109, 95% CI 0.014–0.878; p = 0.037) but not non-PPI ones (adjusted HR: 0.205, 95% CI 0.024–1.745, p = 0.147; Fig. 4). However, no significant effects of PPIs use were observed on the all-cause mortality, non-fatal MI or revascularization (Figs. 3a–c, 4). Particularly, GI bleeding was also not significantly different among these groups regardless of the PPIs use (Table 4, Fig. 3e).
Table 4
Clinical outcomes after 18-month follow-up in sustained PPI, intermittent PPI and non-PPI users
 
Sustained PPI users
Intermittent PPI users
Non-PPI users
p value
Death
4 (2.0%)
6 (3.2%)
9 (3.6%)
0.589
 Fatal MI
0 (0.5%)
1 (0.5%)
1 (0.4%)
0.612
 Stroke
1 (0.5%)
1 (0.5%)
1 (0.4%)
0.978
 Other cardiovascular
1 (0.5%)
1 (0.5%)
1 (0.4%)
0.978
 Non-cardiovascular
0 (0.5%)
0 (0.0%)
4 (1.6%)
0.043
 Unknown
2 (1.0%)
3 (1.6%)
2 (0.8%)
0.723
Rehospitalization
101 (50.2%)
89 (47.3%)
80 (32.1%)
< 0.001
 MI
0 (0.0%)
3 (1.6%)
3 (1.2%)
0.227
 Angina
46 (22.9%)
26 (13.8%)
29 (11.6%)
0.003
 Stroke
3 (1.5%)
1 (0.5%)
5 (2.0%)
0.429
 GI bleeding
1 (0.5%)
0 (0.0%)
0 (0.0%)
0.337
 Other
51 (25.3%)
59 (47.9%)
43 (17.3%)
0.002
Cardiovascular events
    
 Non-fatal MI
1 (0.5%)
3 (1.6%)
2 (0.8%)
0.512
 Revascularization
11 (5.5%)
3 (1.6%)
6 (2.4%)
0.063
 Stroke
10 (5.0%)
1 (0.5%)
9 (3.6%)
0.036
 GI bleeding
2 (1.0%)
0 (0.0%)
2 (0.8%)
0.417
 Other bleeding
0 (0.0%)
0 (0.0%)
0 (0.0%)
GI, gastrointestinal; MI, myocardial infarction; PPI, proton pump inhibitor

The association between PPIs use and 18-month MACE

Beside analyzing the association between PPI use and the individual clinical outcome, we also compared the rate of MACE among three groups. Patients with intermittent PPI use tended to suffer the lowest rate of MACE while sustained users had the highest rate (6.8% for non-PPI users, 5.3% for intermittent users, and 11.4% for sustained ones) (Fig. 5a) but the tendency was nonsignificant (log rank p = 0.056, Fig. 5b). In multivariable Cox regression analysis, intermittent use of PPI was associated with lower risk of MACE than the sustained PPI users (sustained PPI users vs. non-PPI users: adjusted HR: 1.678, 95% CI 0.822–3.423; p = 0.155; intermittent PPI users vs. non-PPI users: adjusted HR: 0.492, 95% CI 0.194–1.248; p = 0.135; intermittent PPI users vs. sustained PPI users: adjusted HR: 0.293, 95% CI 0.119–0.722; p = 0.008).

The association between PPIs use and 18-month NACE

In addition, NACE, including all adverse events, were also estimated. The rates of NACE among three groups were nonsignificant (log rank p = 0.098, Fig. 6). Multivariable Cox regression analysis further showed that intermittent use of PPIs was associated with lower risk of NACE compared with the sustained PPI users, but not non-PPI users (Table 5). To further explore the possible risk factors affecting NACE, subgroup analysis was conducted. As shown in Table 5, intermittent use of PPIs was associated with lower risk of NACE than the other two groups for patients with PCI. Besides, intermittent PPI male users with age over 60 years old, hypertension, CKD and CYP2C19 IM type also suffered less risk of NACE compared with sustained PPI users (Table 5). No different risk ratios of NACE were detected in the subgroups of patients with or without gastroduodenal ulcer (p > 0.05; Table 5). Neither types of the PPIs including pantoprazole exerted beneficial effects on MACE or NACE (Table 5 and Additional file 1: Table S1).
Table 5
Subgroup analysis of NACE in sustained PPI, intermittent PPI users and non-PPI users
Subgroup
No. of events (%)
Adjusted hazard ratio (95% confidence interval), p value
Sustained PPI users
Intermittent PPI users
Non-PPI users
Sustained versus non
Intermittent versus non
Intermittent versus sustained
Overall
27 (13.4)
13 (6.9)
25 (10.0)
1.309 (0.687, 2.495), p = 0.413
0.467 (0.213, 1.025), p = 0.058
0.357 (0.162, 0.786), p = 0.011
Age
      
 ≤ 60 y
3 (5.2)
4 (5.1)
6 (7.3)
1.152 (0.128, 10.388), p = 0.899
0.534 (0.055, 5.155), p = 0.588
0.464 (0.039, 5.486), p = 0.542
 > 60 y
24 (16.8)
9 (8.2)
19 (11.4)
1.392 (0.692, 2.802), p = 0.353
0.440 (0.170, 1.138), p = 0.090
0.316 (0.122, 0.818), p = 0.018
Gender
      
 Male
21 (17.1)
8 (6.9)
13 (7.5)
2.079 (0.937, 4.611), p = 0.072
0.526 (0.193, 1.434), p = 0.209
0.253 (0.097, 0.664), p = 0.005
 Female
6 (7.7)
5 (6.9)
12 (16.0)
0.411 (0.113, 1.489), p = 0.176
0.380 (0.097, 1.496), p = 0.166
0.925 (0.198, 4.317), p = 0.921
Hypertension
      
 Yes
23 (16.0)
9 (7.0)
20 (11.2)
1.234 (0.617, 2.466), p = 0.552
0.360 (0.135, 0.964), p = 0.042*
0.292 (0.109, 0.785), p = 0.015
 No
4 (7.0)
4 (6.7)
5 (7.1)
0.958 (0.140, 6.574), p = 0.965
0.328 (0.040, 2.725), p = 0.302
0.343 (0.038, 3.079), p = 0.339
Stroke
      
 Yes
7 (20.6)
2 (10.5)
12 (25.5)
0.668 (0.206, 2.164), p = 0.501
0.306 (0.031, 3.025), p = 0.311
0.457 (0.047, 4.469), p = 0.501
 No
20 (12.0)
11 (6.5)
13 (6.4)
2.433 (1.020, 5.805), p = 0.045
0.649 (0.258, 1.631), p = 0.358
0.267 (0.104, 0.686), p = 0.066
DM
      
 Yes
8 (18.6)
8 (13.1)
11 (13.8)
2.080 (0.647, 6.692), p = 0.219
0.707 (0.233, 2.151), p = 0.542
0.340 (0.092, 1.257), p = 0.106
 No
19 (12.0)
5 (3.9)
14 (8.3)
1.017 (0.465, 2.224), p = 0.966
0.369 (0.115, 1.185), p = 0.094
0.362 (0.118, 1.117), p = 0.077
CKD
      
 Yes
25 (15.9)
12 (8.1)
25 (14.2)
1.220 (0.633, 2.351), p = 0.552
0.441 (0.195, 0.999), p = 0.050
0.361 (0.157, 0.833), p = 0.017
 No
2 (4.5)
1 (2.5)
0 (0.0)
Gastroduodenal ulcer
      
 Yes
4 (9.1)
1 (3.6)
5 (26.3)
0.485 (0.048, 4.857), p = 0.538
0.191 (0.014, 2.550), p = 0.210
0.393 (0.034, 4.527), p = 0.454
 No
23 (14.7)
12 (7.5)
20 (8.7)
1.740 (0.856, 3.537), p = 0.126
0.583 (0.248, 1.369), p = 0.215
0.335 (0.142, 0.789), p = 0.012
PPI types
      
 Pantoprazole
24 (16.8)
8 (7.2)
25 (10.0)
1.176 (0.508, 2.718), p = 0.705
0.505 (0.196, 1.303), p = 0.158
0.430 (0.150, 1.230), p = 0.115
 Others
3 (5.2)
5 (6.4)
25 (10.0)
1.127 (0.514, 2.471), p = 0.766
0.381 (0.121, 1.202), p = 0.100
0.338 (0.098, 1.169), p = 0.087
PCI
      
 Yes
18 (11.9)
3 (2.5)
14 (9.3)
1.166 (0.522, 2.602), p = 0.708
0.194 (0.052, 0.731), p = 0.015
0.167 (0.045, 0.617), p = 0.007
 No
9 (18.0)
10 (14.7)
11 (11.1)
1.595 (0.532, 4.783), p = 0.405
0.954 (0.341, 2.671), p = 0.929
0.598 (0.178, 2.014), p = 0.407
CYP2C19 genotypes
      
 EM
5 (14.3)
2 (4.5)
5 (10.2)
4.173 (0.606, 28.750), p = 0.147
0.624 (0.088, 4.402), p = 0.636
0.149 (0.022, 1.031), p = 0.054
 IM
9 (16.4)
4 (8.0)
6 (12.8)
3.652 (0.916, 14.559), p = 0.066
0.664 (0.156, 2.827), p = 0.580
0.182 (0.044, 0.751), p = 0.019
 PM
2 (3.6)
1 (6.3)
2 (10.5)
0.058 (0.002, 1.887), p = 0.109
CKD, chronic kidney disease; DM, diabetes mellitus; EM, extensive metabolizers; GI, gastrointestinal; IM, intermediate metabolizers; MI, myocardial infarction; NACE, net adverse clinical events; PCI, percutaneous coronary intervention; PM, poor metabolizers; PPI, proton pump inhibitor

Discussion

The results of this study suggest that in patients with CHD taking clopidogrel, intermittent use of PPIs did not increase the risk of all-cause death, cardiovascular adverse events, and GI bleeding after 18-month follow-up. Instead, compared with the sustained PPI users, intermittent use of PPIs was associated with a reduced risk of NACE after 18 months, especially for male CHD patients with an age over 60 years old after PCI, with hypertension or CKD.
Clopidogrel, often combined with aspirin, is a gold standard treatment for CHD, especially for patients after PCI, to reduce the incidence of cardiovascular adverse events [1820]. However, since clopidogrel needs to be metabolized by the liver CYP450 enzyme system, combined use of the drug metabolized by this enzyme may exert inhibitory effect on clopidogrel [21]. One of the most concerned medications is PPIs, which are often used to prevent GI bleeding [22]. Previous pharmacodynamic studies have shown that the platelet inhibitory rate decreased when clopidogrel was combined with PPIs [2326]. Most observational studies found that patients under both clopidogrel and PPIs therapy had an increased risk of ischemic cardiovascular events [4, 9, 10, 2729]. In the BASKET study, the results of subgroup analysis showed that patients who took PPIs had a higher incidence of NACEs and MI after 3 years than those who did not [10]. The following CAPRIE study confirmed that patients with PPIs suffered a higher risk of ischemic cardiovascular events after one year than non-PPI users [9]. The results of the ADAPT-DES study further proved that the combination of PPIs was associated with increased platelet resistance, and that the incidence of NACE was significantly increased after 2 years [4]. These results indicated that PPIs were able to affect its antiplatelet activity of clopidogrel by disturbing its metabolism, and thereby led to an increased risk of cardiovascular adverse events. However, the results of randomized controlled trials (RCTs) and biased clinical studies were inconsistent [7, 11, 30, 31]. Results from PRINCIPLE-TIMI 44 and TRITON-TIMI 38 studies suggested that clopidogrel and PPIs use had no effects on the clinical adverse events, although the platelet activity induced by clopidogrel was affected by the concurrent use of PPIs [7]. The subsequent COGENT study included a multicenter randomized double-blind case–control study involving 5000 patients with ACS or stent implantation and randomly assigned to omeprazole based on DAPT. After 180 days of follow-up, studies confirmed that the incidence of total cardiovascular death, MI, target vessel revascularization and stroke was not significantly different in both groups when compared to placebo group [12]. Another recent RCT, the PRODIGY study, showed the similar results [11]. Studies in Asian populations have found that clopidogrel combined with PPIs was not associated with cardiovascular events (such as AMI, cardiovascular death, etc.) [32, 33]. The above series of clinical research conclusions were consistent with the results of this study. In this study, compared with patients without use of PPIs, intermittent or sustained use of PPIs did not significantly increase the occurrence of all-cause deaths and adverse clinical events. This further confirmed that although the combined use of PPIs might reduce the antiplatelet function caused by clopidogrel, it did not necessarily indicate an increased risk of clinical adverse events after combined use [34, 35]. Indeed, a recent clinical study involving more than 60,000 patients with gastroesophageal reflux disease suggested that patients taking PPIs were 1.2 times more likely to develop MI than non-PPI users, and their risk of cardiovascular death was twice higher than those who did not take it, but all these were independent of clopidogrel use [29]. The latest meta-analysis of this issue did not find a definitive answer either [34, 35]. Therefore, the clinical prognosis of CHD patients with combined use of clopidogrel and PPIs still requires further confirmation by the well-designed RCTs.
The purpose of using PPIs is mainly to prevent or reduce the occurrence of GI bleeding. Although relatively few studies focused on the benefit of clopidogrel combined with PPIs on GI bleeding, the risk of GI bleeding was found to be reduced after combined with PPIs [11, 12, 31, 36]. In the COGENT study, although there was no significant difference in the overall incidence of cardiovascular adverse events, the risk of GI bleeding was lower in the omeprazole group than in the placebo after 180 days. However, this study was terminated early and the follow-up time was short. Therefore, the risk of GI bleeding after long-term treatment could not be evaluated in this study [12, 31]. The PRODIGY study found that the occurrences of BARC bleeding standard type 2, 3, and 5 bleeding events at 6 months and 24 months were not statistically different between groups with or without PPIs use. However, no comparison among various bleeding events such as GI bleeding events was conducted in that study [11]. This study focused on the observation of GI bleeding in patients with CHD taking clopidogrel combined with PPIs. There was no significant difference in GI bleeding incidence. However, among the sustained PPI users, 45 people suffered gastrointestinal ulcer disease, and the incidence of GI bleeding was 4.4% after 18 months; among the non-PPI users, 20 people suffered stomach intestinal ulcer disease, the incidence of GI bleeding was up to 10% after 18 months. It seemed that the occurrence of GI bleeding in the non-PPI users was higher than that in the sustained PPI users. Due to the small number of events, it could not be confirmed by multivariate analysis. However, this result was consistent with the research published earlier by the Chitose T. team in Japan, which suggested that the group without the PPIs use showed a tendency of increasing GI bleeding [36]. Therefore, use of PPIs was unlikely to increase the risk of clinical adverse events but may exert benefit on GI bleeding prevention.
How to prescribe PPIs to the CHD patients in clinical practice is another important issue. Till now, only few studies have explored the impact of various frequencies of PPIs use on clinical prognosis. Such as the PRODIGY study, the study defined those who took PPIs at the 30-day follow-up as the PPIs use group. Patients who took less than 30-day PPIs and those who discontinued PPIs were excluded [11]. Other studies, such as ADAPT-DES and BASKET, regarded patients discharged with PPIs as the PPI group, but did not consider the changes of PPIs dosage or the frequency of concurrent use after discharge [4, 10]. In the clinical practice, most patients may accept PPIs treatment when they suffer digestive discomfort. Therefore, to identify the optimal PPI regimens in CHD patients to achieve beneficial clinical impacts is of great importance. Based on the duration of PPI use during the follow-up period, PPI users were divided into the sustained and intermittent PPI users groups. No significant difference was found on the individual clinical adverse events of the two PPI-user groups. Surprisingly, the intermittent PPI user group had a significantly lower incidence of NACE than the sustained PPI users, of which the main benefit may be due to a reduction in stroke. Further subgroup analysis suggested that male CHD patients with an age over 60 old years, after PCI treatment, with hypertension or CKD, might benefit more from intermittent use of PPIs. Therefore, for certain populations, intermittent use may be related to a reduction in NACE. In fact, this was in line with the recommendations of the latest US ACC/AHA guidelines. The guideline considered that PPIs may be used appropriately for people at high risk, such as the elderly, who were at high risk for GI bleeding [19, 20]. In addition, for PCI patients, who were necessary to take DAPT continuously for at least 1 year, it was particularly critical to prevent complications such as GI bleeding that forced to discontinue DAPT. The present study indicated that the appropriate and intermittent concurrent use of PPI with clopidogrel may be available for patients after PCI. Nevertheless, because of the low incidence of GI bleeding and other bleeding events in this study, it was impossible to further analyze whether the benefit of NACE came from the reduction of GI bleeding or was affected by the imbalanced baseline. However, multivariate analysis adjusted for possible confounders reduced the impact of baseline imbalances to a certain extent. According to previous large-scale clinical studies such as the PRODIGY study, the incidence of NACE in the sustained user and the non-PPI user group in this study was comparable, also between 10.0 and 15.0%, while the intermittent user group was significantly reduced, which indicated that the results of this study had considerable credibility [11].
There were several limitations in this study. Firstly, this study was a single-center retrospective study. Population included is heterogeneous, considering patients in single and dual antiplatelet therapy. The included sample size was relatively small. Thus, the inherent bias would inevitably occur. Secondly, the small number of clinical adverse events might have a certain impact on the research results. Thirdly, the occurrence of clinical events and the consumption of drugs were obtained by telephone or outpatient follow-up, which might inevitably lead to reporting bias. However, the majority of baseline data, medication treatments, and some clinical adverse events were collected or confirmed in the hospital medical system that would minimize the reporting bias. Fourthly, there are only 65 NACEs in this study, while the multivariable model used 11 covariates, which may have a high risk of overfitting. Thus, we should be cautious when interpreting the current findings.

Conclusion

In summary, the intermittent combination of clopidogrel and PPI did not increase the risk of all-cause death and cardiovascular adverse events in patients with CHD. Intermittent use of PPIs is associated with a lower rate of MACE and NACE. Although there was no statistically significant difference in the incidence of GI bleeding among the three groups, there was a tendency of decreased incidence of GI bleeding with the use of PPIs. However, larger RCTs are needed to further confirm the conclusions of this study.

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12872-021-01884-z.

Acknowledgement

None.
This study was approved by Sun Yat-Sen University Ethics Committee. All procedures performed in this study involving human participants were in accordance with the ethical standards. Informed consent was obtained from each participant.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Collaborators GMaCoD. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015;385(9963):117–71.CrossRef Collaborators GMaCoD. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015;385(9963):117–71.CrossRef
2.
Zurück zum Zitat Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, et al. Heart disease and stroke statistics-2017 update: a report from the American Heart Association. Circulation. 2017;135(10):e146–603.CrossRef Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, et al. Heart disease and stroke statistics-2017 update: a report from the American Heart Association. Circulation. 2017;135(10):e146–603.CrossRef
3.
Zurück zum Zitat Townsend N, Wilson L, Bhatnagar P, Wickramasinghe K, Rayner M, Nichols M. Cardiovascular disease in Europe: epidemiological update 2016. Eur Heart J. 2016;37(42):3232–45.CrossRef Townsend N, Wilson L, Bhatnagar P, Wickramasinghe K, Rayner M, Nichols M. Cardiovascular disease in Europe: epidemiological update 2016. Eur Heart J. 2016;37(42):3232–45.CrossRef
4.
Zurück zum Zitat Weisz G, Smilowitz NR, Kirtane AJ, Rinaldi MJ, Parvataneni R, Xu K, Stuckey TD, Maehara A, Witzenbichler B, Neumann FJ, et al. Proton pump inhibitors, platelet reactivity, and cardiovascular outcomes after drug-eluting stents in clopidogrel-treated patients: the ADAPT-DES study. Circ Cardiovasc Interv. 2015;8(10):e001952.CrossRef Weisz G, Smilowitz NR, Kirtane AJ, Rinaldi MJ, Parvataneni R, Xu K, Stuckey TD, Maehara A, Witzenbichler B, Neumann FJ, et al. Proton pump inhibitors, platelet reactivity, and cardiovascular outcomes after drug-eluting stents in clopidogrel-treated patients: the ADAPT-DES study. Circ Cardiovasc Interv. 2015;8(10):e001952.CrossRef
5.
Zurück zum Zitat Sibbing D, Morath T, Stegherr J, Braun S, Vogt W, Hadamitzky M, Schomig A, Kastrati A, von Beckerath N. Impact of proton pump inhibitors on the antiplatelet effects of clopidogrel. Thromb Haemost. 2009;101(4):714–9.CrossRef Sibbing D, Morath T, Stegherr J, Braun S, Vogt W, Hadamitzky M, Schomig A, Kastrati A, von Beckerath N. Impact of proton pump inhibitors on the antiplatelet effects of clopidogrel. Thromb Haemost. 2009;101(4):714–9.CrossRef
6.
Zurück zum Zitat Small DS, Farid NA, Payne CD, Weerakkody GJ, Li YG, Brandt JT, Salazar DE, Winters KJ. Effects of the proton pump inhibitor lansoprazole on the pharmacokinetics and pharmacodynamics of prasugrel and clopidogrel. J Clin Pharmacol. 2008;48(4):475–84.CrossRef Small DS, Farid NA, Payne CD, Weerakkody GJ, Li YG, Brandt JT, Salazar DE, Winters KJ. Effects of the proton pump inhibitor lansoprazole on the pharmacokinetics and pharmacodynamics of prasugrel and clopidogrel. J Clin Pharmacol. 2008;48(4):475–84.CrossRef
7.
Zurück zum Zitat O’Donoghue ML, Braunwald E, Antman EM, Murphy SA, Bates ER, Rozenman Y, Michelson AD, Hautvast RW, Ver Lee PN, Close SL, et al. Pharmacodynamic effect and clinical efficacy of clopidogrel and prasugrel with or without a proton-pump inhibitor: an analysis of two randomised trials. Lancet (London, England). 2009;374(9694):989–97.CrossRef O’Donoghue ML, Braunwald E, Antman EM, Murphy SA, Bates ER, Rozenman Y, Michelson AD, Hautvast RW, Ver Lee PN, Close SL, et al. Pharmacodynamic effect and clinical efficacy of clopidogrel and prasugrel with or without a proton-pump inhibitor: an analysis of two randomised trials. Lancet (London, England). 2009;374(9694):989–97.CrossRef
8.
Zurück zum Zitat Gilard M, Arnaud B, Cornily JC, Le Gal G, Lacut K, Le Calvez G, Mansourati J, Mottier D, Abgrall JF, Boschat J. Influence of omeprazole on the antiplatelet action of clopidogrel associated with aspirin: the randomized, double-blind OCLA (Omeprazole CLopidogrel Aspirin) study. J Am Coll Cardiol. 2008;51(3):256–60.CrossRef Gilard M, Arnaud B, Cornily JC, Le Gal G, Lacut K, Le Calvez G, Mansourati J, Mottier D, Abgrall JF, Boschat J. Influence of omeprazole on the antiplatelet action of clopidogrel associated with aspirin: the randomized, double-blind OCLA (Omeprazole CLopidogrel Aspirin) study. J Am Coll Cardiol. 2008;51(3):256–60.CrossRef
9.
Zurück zum Zitat Dunn SP, Steinhubl SR, Bauer D, Charnigo RJ, Berger PB, Topol EJ. Impact of proton pump inhibitor therapy on the efficacy of clopidogrel in the CAPRIE and CREDO trials. J Am Heart Assoc. 2013;2(1):e004564.CrossRef Dunn SP, Steinhubl SR, Bauer D, Charnigo RJ, Berger PB, Topol EJ. Impact of proton pump inhibitor therapy on the efficacy of clopidogrel in the CAPRIE and CREDO trials. J Am Heart Assoc. 2013;2(1):e004564.CrossRef
10.
Zurück zum Zitat Burkard T, Kaiser CA, Brunner-La Rocca H, Osswald S, Pfisterer ME, Jeger RV. Combined clopidogrel and proton pump inhibitor therapy is associated with higher cardiovascular event rates after percutaneous coronary intervention: a report from the BASKET trial. J Intern Med. 2012;271(3):257–63.CrossRef Burkard T, Kaiser CA, Brunner-La Rocca H, Osswald S, Pfisterer ME, Jeger RV. Combined clopidogrel and proton pump inhibitor therapy is associated with higher cardiovascular event rates after percutaneous coronary intervention: a report from the BASKET trial. J Intern Med. 2012;271(3):257–63.CrossRef
11.
Zurück zum Zitat Gargiulo G, Costa F, Ariotti S, Biscaglia S, Campo G, Esposito G, Leonardi S, Vranckx P, Windecker S, Valgimigli M. Impact of proton pump inhibitors on clinical outcomes in patients treated with a 6- or 24-month dual-antiplatelet therapy duration: Insights from the PROlonging Dual-antiplatelet treatment after Grading stent-induced Intimal hyperplasia studY trial. Am Heart J. 2016;174:95–102.CrossRef Gargiulo G, Costa F, Ariotti S, Biscaglia S, Campo G, Esposito G, Leonardi S, Vranckx P, Windecker S, Valgimigli M. Impact of proton pump inhibitors on clinical outcomes in patients treated with a 6- or 24-month dual-antiplatelet therapy duration: Insights from the PROlonging Dual-antiplatelet treatment after Grading stent-induced Intimal hyperplasia studY trial. Am Heart J. 2016;174:95–102.CrossRef
12.
Zurück zum Zitat Bhatt DL, Cryer BL, Contant CF, Cohen M, Lanas A, Schnitzer TJ, Shook TL, Lapuerta P, Goldsmith MA, Laine L, et al. Clopidogrel with or without omeprazole in coronary artery disease. N Engl J Med. 2010;363(20):1909–17.CrossRef Bhatt DL, Cryer BL, Contant CF, Cohen M, Lanas A, Schnitzer TJ, Shook TL, Lapuerta P, Goldsmith MA, Laine L, et al. Clopidogrel with or without omeprazole in coronary artery disease. N Engl J Med. 2010;363(20):1909–17.CrossRef
13.
Zurück zum Zitat Xie HG, Zou JJ, Hu ZY, Zhang JJ, Ye F, Chen SL. Individual variability in the disposition of and response to clopidogrel: pharmacogenomics and beyond. Pharmacol Ther. 2011;129(3):267–89.CrossRef Xie HG, Zou JJ, Hu ZY, Zhang JJ, Ye F, Chen SL. Individual variability in the disposition of and response to clopidogrel: pharmacogenomics and beyond. Pharmacol Ther. 2011;129(3):267–89.CrossRef
14.
Zurück zum Zitat Xie HG, Kim RB, Wood AJ, Stein CM. Molecular basis of ethnic differences in drug disposition and response. Ann Rev Pharmacol Toxicol. 2001;41:815–50.CrossRef Xie HG, Kim RB, Wood AJ, Stein CM. Molecular basis of ethnic differences in drug disposition and response. Ann Rev Pharmacol Toxicol. 2001;41:815–50.CrossRef
15.
Zurück zum Zitat Rosemary J, Adithan C. The pharmacogenetics of CYP2C9 and CYP2C19: ethnic variation and clinical significance. Curr Clin Pharmacol. 2007;2(1):93–109.CrossRef Rosemary J, Adithan C. The pharmacogenetics of CYP2C9 and CYP2C19: ethnic variation and clinical significance. Curr Clin Pharmacol. 2007;2(1):93–109.CrossRef
16.
Zurück zum Zitat Li-Wan-Po A, Girard T, Farndon P, Cooley C, Lithgow J. Pharmacogenetics of CYP2C19: functional and clinical implications of a new variant CYP2C19*17. Br J Clin Pharmacol. 2010;69(3):222–30.CrossRef Li-Wan-Po A, Girard T, Farndon P, Cooley C, Lithgow J. Pharmacogenetics of CYP2C19: functional and clinical implications of a new variant CYP2C19*17. Br J Clin Pharmacol. 2010;69(3):222–30.CrossRef
17.
Zurück zum Zitat Mehran R, Rao SV, Bhatt DL, Gibson CM, Caixeta A, Eikelboom J, Kaul S, Wiviott SD, Menon V, Nikolsky E, et al. Standardized bleeding definitions for cardiovascular clinical trials: a consensus report from the Bleeding Academic Research Consortium. Circulation. 2011;123(23):2736–47.CrossRef Mehran R, Rao SV, Bhatt DL, Gibson CM, Caixeta A, Eikelboom J, Kaul S, Wiviott SD, Menon V, Nikolsky E, et al. Standardized bleeding definitions for cardiovascular clinical trials: a consensus report from the Bleeding Academic Research Consortium. Circulation. 2011;123(23):2736–47.CrossRef
18.
Zurück zum Zitat Roffi M, Patrono C, Collet JP, Mueller C, Valgimigli M, Andreotti F, Bax JJ, Borger MA, Brotons C, Chew DP, et al. 2015 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: Task Force for the Management of Acute Coronary Syndromes in Patients Presenting without Persistent ST-Segment Elevation of the European Society of Cardiology (ESC). Eur Heart J. 2016;37(3):267–315.CrossRef Roffi M, Patrono C, Collet JP, Mueller C, Valgimigli M, Andreotti F, Bax JJ, Borger MA, Brotons C, Chew DP, et al. 2015 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: Task Force for the Management of Acute Coronary Syndromes in Patients Presenting without Persistent ST-Segment Elevation of the European Society of Cardiology (ESC). Eur Heart J. 2016;37(3):267–315.CrossRef
19.
Zurück zum Zitat Levine GN, Bates ER, Blankenship JC, Bailey SR, Bittl JA, Cercek B, Chambers CE, Ellis SG, Guyton RA, Hollenberg SM, et al. 2015 ACC/AHA/SCAI focused update on primary percutaneous coronary intervention for patients with ST-elevation myocardial infarction: an update of the 2011 ACCF/AHA/SCAI guideline for percutaneous coronary intervention and the 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction. J Am Coll Cardiol. 2016;67(10):1235–50.CrossRef Levine GN, Bates ER, Blankenship JC, Bailey SR, Bittl JA, Cercek B, Chambers CE, Ellis SG, Guyton RA, Hollenberg SM, et al. 2015 ACC/AHA/SCAI focused update on primary percutaneous coronary intervention for patients with ST-elevation myocardial infarction: an update of the 2011 ACCF/AHA/SCAI guideline for percutaneous coronary intervention and the 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction. J Am Coll Cardiol. 2016;67(10):1235–50.CrossRef
20.
Zurück zum Zitat Levine GN, Bates ER, Bittl JA, Brindis RG, Fihn SD, Fleisher LA, Granger CB, Lange RA, Mack MJ, Mauri L, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: a report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines: an update of the 2011 ACCF/AHA/SCAI guideline for percutaneous coronary intervention, 2011 ACCF/AHA guideline for coronary artery bypass graft surgery, 2012 ACC/AHA/ACP/AATS/PCNA/SCAI/STS guideline for the diagnosis and management of patients with stable ischemic heart disease, 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction, 2014 AHA/ACC guideline for the management of patients with non-ST-elevation acute coronary syndromes, and 2014 ACC/AHA guideline on perioperative cardiovascular evaluation and management of patients undergoing noncardiac surgery. Circulation. 2016;134(10):e123-155.CrossRef Levine GN, Bates ER, Bittl JA, Brindis RG, Fihn SD, Fleisher LA, Granger CB, Lange RA, Mack MJ, Mauri L, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: a report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines: an update of the 2011 ACCF/AHA/SCAI guideline for percutaneous coronary intervention, 2011 ACCF/AHA guideline for coronary artery bypass graft surgery, 2012 ACC/AHA/ACP/AATS/PCNA/SCAI/STS guideline for the diagnosis and management of patients with stable ischemic heart disease, 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction, 2014 AHA/ACC guideline for the management of patients with non-ST-elevation acute coronary syndromes, and 2014 ACC/AHA guideline on perioperative cardiovascular evaluation and management of patients undergoing noncardiac surgery. Circulation. 2016;134(10):e123-155.CrossRef
21.
Zurück zum Zitat Jiang XL, Samant S, Lesko LJ, Schmidt S. Clinical pharmacokinetics and pharmacodynamics of clopidogrel. Clin Pharmacokinet. 2015;54(2):147–66.CrossRef Jiang XL, Samant S, Lesko LJ, Schmidt S. Clinical pharmacokinetics and pharmacodynamics of clopidogrel. Clin Pharmacokinet. 2015;54(2):147–66.CrossRef
22.
Zurück zum Zitat Harrison RW, Mahaffey KW. Clopidogrel and PPI interaction: clinically relevant or not? Curr Cardiol Rep. 2012;14(1):49–58.CrossRef Harrison RW, Mahaffey KW. Clopidogrel and PPI interaction: clinically relevant or not? Curr Cardiol Rep. 2012;14(1):49–58.CrossRef
23.
Zurück zum Zitat Angiolillo DJ, Gibson CM, Cheng S, Ollier C, Nicolas O, Bergougnan L, Perrin L, LaCreta FP, Hurbin F, Dubar M. Differential effects of omeprazole and pantoprazole on the pharmacodynamics and pharmacokinetics of clopidogrel in healthy subjects: randomized, placebo-controlled, crossover comparison studies. Clin Pharmacol Ther. 2011;89(1):65–74.CrossRef Angiolillo DJ, Gibson CM, Cheng S, Ollier C, Nicolas O, Bergougnan L, Perrin L, LaCreta FP, Hurbin F, Dubar M. Differential effects of omeprazole and pantoprazole on the pharmacodynamics and pharmacokinetics of clopidogrel in healthy subjects: randomized, placebo-controlled, crossover comparison studies. Clin Pharmacol Ther. 2011;89(1):65–74.CrossRef
24.
Zurück zum Zitat Fontes-Carvalho R, Albuquerque A, Araujo C, Pimentel-Nunes P, Ribeiro VG. Omeprazole, but not pantoprazole, reduces the antiplatelet effect of clopidogrel: a randomized clinical crossover trial in patients after myocardial infarction evaluating the clopidogrel-PPIs drug interaction. Eur J Gastroenterol Hepatol. 2011;23(5):396–404.CrossRef Fontes-Carvalho R, Albuquerque A, Araujo C, Pimentel-Nunes P, Ribeiro VG. Omeprazole, but not pantoprazole, reduces the antiplatelet effect of clopidogrel: a randomized clinical crossover trial in patients after myocardial infarction evaluating the clopidogrel-PPIs drug interaction. Eur J Gastroenterol Hepatol. 2011;23(5):396–404.CrossRef
25.
Zurück zum Zitat Frelinger AL III, Lee RD, Mulford DJ, Wu J, Nudurupati S, Nigam A, Brooks JK, Bhatt DL, Michelson AD. A randomized, 2-period, crossover design study to assess the effects of dexlansoprazole, lansoprazole, esomeprazole, and omeprazole on the steady-state pharmacokinetics and pharmacodynamics of clopidogrel in healthy volunteers. J Am Coll Cardiol. 2012;59(14):1304–11.CrossRef Frelinger AL III, Lee RD, Mulford DJ, Wu J, Nudurupati S, Nigam A, Brooks JK, Bhatt DL, Michelson AD. A randomized, 2-period, crossover design study to assess the effects of dexlansoprazole, lansoprazole, esomeprazole, and omeprazole on the steady-state pharmacokinetics and pharmacodynamics of clopidogrel in healthy volunteers. J Am Coll Cardiol. 2012;59(14):1304–11.CrossRef
26.
Zurück zum Zitat Furuta T, Iwaki T, Umemura K. Influences of different proton pump inhibitors on the anti-platelet function of clopidogrel in relation to CYP2C19 genotypes. Br J Clin Pharmacol. 2010;70(3):383–92.CrossRef Furuta T, Iwaki T, Umemura K. Influences of different proton pump inhibitors on the anti-platelet function of clopidogrel in relation to CYP2C19 genotypes. Br J Clin Pharmacol. 2010;70(3):383–92.CrossRef
27.
Zurück zum Zitat Maggio M, Corsonello A, Ceda GP, Cattabiani C, Lauretani F, Butto V, Ferrucci L, Bandinelli S, Abbatecola AM, Spazzafumo L, et al. Proton pump inhibitors and risk of 1-year mortality and rehospitalization in older patients discharged from acute care hospitals. JAMA Intern Med. 2013;173(7):518–23.CrossRef Maggio M, Corsonello A, Ceda GP, Cattabiani C, Lauretani F, Butto V, Ferrucci L, Bandinelli S, Abbatecola AM, Spazzafumo L, et al. Proton pump inhibitors and risk of 1-year mortality and rehospitalization in older patients discharged from acute care hospitals. JAMA Intern Med. 2013;173(7):518–23.CrossRef
28.
Zurück zum Zitat Charlot M, Ahlehoff O, Norgaard ML, Jorgensen CH, Sorensen R, Abildstrom SZ, Hansen PR, Madsen JK, Kober L, Torp-Pedersen C, et al. Proton-pump inhibitors are associated with increased cardiovascular risk independent of clopidogrel use: a nationwide cohort study. Ann Intern Med. 2010;153(6):378–86.CrossRef Charlot M, Ahlehoff O, Norgaard ML, Jorgensen CH, Sorensen R, Abildstrom SZ, Hansen PR, Madsen JK, Kober L, Torp-Pedersen C, et al. Proton-pump inhibitors are associated with increased cardiovascular risk independent of clopidogrel use: a nationwide cohort study. Ann Intern Med. 2010;153(6):378–86.CrossRef
29.
Zurück zum Zitat Shah NH, LePendu P, Bauer-Mehren A, Ghebremariam YT, Iyer SV, Marcus J, Nead KT, Cooke JP, Leeper NJ. Proton pump inhibitor usage and the risk of myocardial infarction in the general population. PLoS ONE. 2015;10(6):e0124653.CrossRef Shah NH, LePendu P, Bauer-Mehren A, Ghebremariam YT, Iyer SV, Marcus J, Nead KT, Cooke JP, Leeper NJ. Proton pump inhibitor usage and the risk of myocardial infarction in the general population. PLoS ONE. 2015;10(6):e0124653.CrossRef
30.
Zurück zum Zitat Simon T, Steg PG, Gilard M, Blanchard D, Bonello L, Hanssen M, Lardoux H, Coste P, Lefevre T, Drouet E, et al. Clinical events as a function of proton pump inhibitor use, clopidogrel use, and cytochrome P450 2C19 genotype in a large nationwide cohort of acute myocardial infarction: results from the French Registry of Acute ST-Elevation and Non-ST-Elevation Myocardial Infarction (FAST-MI) registry. Circulation. 2011;123(5):474–82.CrossRef Simon T, Steg PG, Gilard M, Blanchard D, Bonello L, Hanssen M, Lardoux H, Coste P, Lefevre T, Drouet E, et al. Clinical events as a function of proton pump inhibitor use, clopidogrel use, and cytochrome P450 2C19 genotype in a large nationwide cohort of acute myocardial infarction: results from the French Registry of Acute ST-Elevation and Non-ST-Elevation Myocardial Infarction (FAST-MI) registry. Circulation. 2011;123(5):474–82.CrossRef
31.
Zurück zum Zitat Vaduganathan M, Bhatt DL, Cryer BL, Liu Y, Hsieh WH, Doros G, Cohen M, Lanas A, Schnitzer TJ, Shook TL, et al. Proton-pump inhibitors reduce gastrointestinal events regardless of aspirin dose in patients requiring dual antiplatelet therapy. J Am Coll Cardiol. 2016;67(14):1661–71.CrossRef Vaduganathan M, Bhatt DL, Cryer BL, Liu Y, Hsieh WH, Doros G, Cohen M, Lanas A, Schnitzer TJ, Shook TL, et al. Proton-pump inhibitors reduce gastrointestinal events regardless of aspirin dose in patients requiring dual antiplatelet therapy. J Am Coll Cardiol. 2016;67(14):1661–71.CrossRef
32.
Zurück zum Zitat Shih CJ, Chen YT, Ou SM, Li SY, Chen TJ, Wang SJ. Proton pump inhibitor use represents an independent risk factor for myocardial infarction. Int J Cardiol. 2014;177(1):292–7.CrossRef Shih CJ, Chen YT, Ou SM, Li SY, Chen TJ, Wang SJ. Proton pump inhibitor use represents an independent risk factor for myocardial infarction. Int J Cardiol. 2014;177(1):292–7.CrossRef
33.
Zurück zum Zitat Zou JJ, Chen SL, Tan J, Lin L, Zhao YY, Xu HM, Lin S, Zhang J, Fan HW, Xie HG. Increased risk for developing major adverse cardiovascular events in stented Chinese patients treated with dual antiplatelet therapy after concomitant use of the proton pump inhibitor. PLoS ONE. 2014;9(1):e84985.CrossRef Zou JJ, Chen SL, Tan J, Lin L, Zhao YY, Xu HM, Lin S, Zhang J, Fan HW, Xie HG. Increased risk for developing major adverse cardiovascular events in stented Chinese patients treated with dual antiplatelet therapy after concomitant use of the proton pump inhibitor. PLoS ONE. 2014;9(1):e84985.CrossRef
34.
Zurück zum Zitat Cardoso RN, Benjo AM, DiNicolantonio JJ, Garcia DC, Macedo FY, El-Hayek G, Nadkarni GN, Gili S, Iannaccone M, Konstantinidis I, et al. Incidence of cardiovascular events and gastrointestinal bleeding in patients receiving clopidogrel with and without proton pump inhibitors: an updated meta-analysis. Open Heart. 2015;2(1):e000248.CrossRef Cardoso RN, Benjo AM, DiNicolantonio JJ, Garcia DC, Macedo FY, El-Hayek G, Nadkarni GN, Gili S, Iannaccone M, Konstantinidis I, et al. Incidence of cardiovascular events and gastrointestinal bleeding in patients receiving clopidogrel with and without proton pump inhibitors: an updated meta-analysis. Open Heart. 2015;2(1):e000248.CrossRef
35.
Zurück zum Zitat Melloni C, Washam JB, Jones WS, Halim SA, Hasselblad V, Mayer SB, Heidenfelder BL, Dolor RJ. Conflicting results between randomized trials and observational studies on the impact of proton pump inhibitors on cardiovascular events when coadministered with dual antiplatelet therapy: systematic review. Circ Cardiovasc Qual Outcomes. 2015;8(1):47–55.CrossRef Melloni C, Washam JB, Jones WS, Halim SA, Hasselblad V, Mayer SB, Heidenfelder BL, Dolor RJ. Conflicting results between randomized trials and observational studies on the impact of proton pump inhibitors on cardiovascular events when coadministered with dual antiplatelet therapy: systematic review. Circ Cardiovasc Qual Outcomes. 2015;8(1):47–55.CrossRef
36.
Zurück zum Zitat Chitose T, Hokimoto S, Oshima S, Nakao K, Fujimoto K, Miyao Y, Shimomura H, Tsunoda R, Maruyama H, Hirose T, et al. Clinical outcomes following coronary stenting in Japanese patients treated with and without proton pump inhibitor. Circ J Off J Jpn Circ Soc. 2012;76(1):71–8. Chitose T, Hokimoto S, Oshima S, Nakao K, Fujimoto K, Miyao Y, Shimomura H, Tsunoda R, Maruyama H, Hirose T, et al. Clinical outcomes following coronary stenting in Japanese patients treated with and without proton pump inhibitor. Circ J Off J Jpn Circ Soc. 2012;76(1):71–8.
Metadaten
Titel
Intermittent concurrent use of clopidogrel and proton pump inhibitors did not increase risk of adverse clinical outcomes in Chinese patients with coronary artery disease
verfasst von
Wanbing He
Xiaorong Shu
Enyi Zhu
Bingqing Deng
Yongqing Lin
Xiaoying Wu
Zenan Zhou
Jingfeng Wang
Ruqiong Nie
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
BMC Cardiovascular Disorders / Ausgabe 1/2021
Elektronische ISSN: 1471-2261
DOI
https://doi.org/10.1186/s12872-021-01884-z

Weitere Artikel der Ausgabe 1/2021

BMC Cardiovascular Disorders 1/2021 Zur Ausgabe

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Medizinstudium Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Vorhofflimmern bei Jüngeren gefährlicher als gedacht

06.05.2024 Vorhofflimmern Nachrichten

Immer mehr jüngere Menschen leiden unter Vorhofflimmern. Betroffene unter 65 Jahren haben viele Risikofaktoren und ein signifikant erhöhtes Sterberisiko verglichen mit Gleichaltrigen ohne die Erkrankung.

Chronisches Koronarsyndrom: Gefahr von Hospitalisierung wegen Herzinsuffizienz

06.05.2024 Herzinsuffizienz Nachrichten

Obwohl ein rezidivierender Herzinfarkt bei chronischem Koronarsyndrom wahrscheinlich die Hauptsorge sowohl der Patienten als auch der Ärzte ist, sind andere Ereignisse womöglich gefährlicher. Laut einer französischen Studie stellt eine Hospitalisation wegen Herzinsuffizienz eine größere Gefahr dar.

Das Risiko für Vorhofflimmern in der Bevölkerung steigt

02.05.2024 Vorhofflimmern Nachrichten

Das Risiko, im Lauf des Lebens an Vorhofflimmern zu erkranken, ist in den vergangenen 20 Jahren gestiegen: Laut dänischen Zahlen wird es drei von zehn Personen treffen. Das hat Folgen weit über die Schlaganfallgefährdung hinaus.

Update Kardiologie

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