901
|
Agarwal N, Mahtta D, Rambarat CA, Elgendy I, Mahmoud AN. Optimum Antithrombotic Therapy in Patients Requiring Long-Term Anticoagulation and Undergoing Percutaneous Coronary Intervention. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5690640. [PMID: 29770334 PMCID: PMC5889881 DOI: 10.1155/2018/5690640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
Abstract
Management of patients on long-term anticoagulation requiring percutaneous coronary intervention is challenging. Triple therapy with oral anticoagulant and dual antiplatelet therapy is the standard of care. However, there is no strong evidence to support this strategy. There is emerging data regarding the safety and efficacy of dual therapy with oral anticoagulant and single antiplatelet therapy in these patients. In this comprehensive review we highlight available evidence regarding various antithrombotic regimens' efficacy and safety in patient with coronary artery disease undergoing percutaneous coronary intervention with long-term anticoagulation therapy requirements.
Collapse
Affiliation(s)
- Nayan Agarwal
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Dhruv Mahtta
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | | | - Islam Elgendy
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Ahmed N. Mahmoud
- Department of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
902
|
Gach O, Nyssen A, Pirlet C, Magne J, Oury C, Lancellotti P. Pretreatment with P2Y12 inhibitors and outcome in patients with ST-segment elevation myocardial infarction treated by primary percutaneous coronary intervention. J Cardiovasc Med (Hagerstown) 2018. [PMID: 29528868 DOI: 10.2459/jcm.0000000000000643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIMS Preload with clopidogrel, ticagrelor, or prasugrel in the setting of ST-segment elevation myocardial infarction (STEMI) treated by primary percutaneous coronary intervention (PCI) is frequently applied. Limited data are available regarding the outcome impact of pretreatment with these drugs in the real world. METHODS AND RESULTS The outcome of 760 STEMI patients treated by primary PCI receiving clopidogrel, prasugrel, or ticagrelor (n = 269, 327, 164, respectively) was evaluated. Patients in the clopidogrel group were older, whereas those in the ticagrelor group had less hypertension but were more active smokers. Angiographic characteristics were comparable among the three groups. At 1 month, more events were observed in the clopidogrel group (11.1%) than in the ticagrelor and prasugrel groups (7.1 vs. 5.1%, P = 0.025), whereas the number of events in the ticagrelor and prasugrel groups did not differ. At 1 year, similar differences existed, mainly driven by a higher rate of death (19.5%, P = 0.008) or stent thrombosis (2 vs. 1.3% for ticagrelor, P = 0.132; vs. 0.3% for prasugrel, P = 0.07) in the clopidogrel group. In-hospital and 1-year bleeding rates were similar between groups. CONCLUSION In real-world practice, pretreatment with prasugrel or ticagrelor in ongoing STEMI treated by primary PCI seems to be a well tolerated alternative strategy compared with clopidogrel but provides superior benefit in terms of outcomes.
Collapse
Affiliation(s)
- Olivier Gach
- Department of Cardiology, GIGA Cardiovascular Sciences, University of Liège Hospital, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Astrid Nyssen
- Department of Cardiology, GIGA Cardiovascular Sciences, University of Liège Hospital, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Charles Pirlet
- Department of Cardiology, GIGA Cardiovascular Sciences, University of Liège Hospital, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Julien Magne
- Centre Hospitalier Universitaire de Limoges, Service de Cardiologie, Limoges, France
| | - Cécile Oury
- Department of Cardiology, GIGA Cardiovascular Sciences, University of Liège Hospital, University of Liège, CHU Sart Tilman, Liège, Belgium
| | - Patrizio Lancellotti
- Department of Cardiology, GIGA Cardiovascular Sciences, University of Liège Hospital, University of Liège, CHU Sart Tilman, Liège, Belgium.,Gruppo Villa Maria Care and Research, Anthea Hospital, Bari, Italy
| |
Collapse
|
903
|
Doshi R, Shah J, Jauhar V, Decter D, Jauhar R, Meraj P. Comparison of drug eluting stents (DESs) and bare metal stents (BMSs) with STEMI: who received BMS in the era of 2nd generation DES? Heart Lung 2018; 47:231-236. [PMID: 29544863 DOI: 10.1016/j.hrtlng.2018.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/09/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND The aim of this study was to analyze the indications for using bare metal stents (BMSs) in hospitalizations with ST segment elevation myocardial infarction (STEMI) undergoing percutaneous coronary intervention (PCI). METHODS The study cohorts were identified from the National Inpatient Sample database from 2010-2014 using appropriate, International Classification of Diseases, 9th Revision, Clinical Modification, diagnostic and procedural codes. RESULTS A total of 123,487 hospitalizations were identified for this study. Drug eluting stent (DES) use demonstrated lower in-hospital mortality (5.8% vs. 3.3%, P = < 0.01) and other in-hospital outcomes, thus resulting in lower hospitalization stay. Higher age, black race, greater comorbidity burden, inferior wall myocardial infarction, and the use of mechanical circulatory devices were all associated with BMS use. CONCLUSION DES was the preferred standard of care in the era of 2nd generation DES; however, BMSs were used in hospitalizations with high-risk procedures and multiple risk factors.
Collapse
Affiliation(s)
- Rajkumar Doshi
- Department of Cardiology, North Shore University Hospital, Northwell Health, Manhasset, New York.
| | - Jay Shah
- Department of Internal Medicine, Mercy Saint Vincent Hospital, University of Toledo, Toledo, OH
| | - Varun Jauhar
- Department of Cardiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| | - Dean Decter
- Department of Cardiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| | - Rajiv Jauhar
- Department of Cardiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| | - Perwaiz Meraj
- Department of Cardiology, North Shore University Hospital, Northwell Health, Manhasset, New York
| |
Collapse
|
904
|
Ananthakrishna R, Kristanto W, Liu L, Chan SP, Loh PH, Tay EL, Chan KH, Chan MY, Lee CH, Low AF, Tan HC, Loh JP. Incidence and predictors of target lesion failure in a multiethnic Asian population receiving the SYNERGY coronary stent: A prospective all-comers registry. Catheter Cardiovasc Interv 2018. [PMID: 29513378 DOI: 10.1002/ccd.27577] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVES To evaluate the target lesion failure (TLF) rate of the SYNERGY stent in all-comers, multiethnic Asian population. BACKGROUND Currently, most drug eluting stents deliver anti-proliferative drugs from a durable polymer which is associated with a risk of late stent thrombosis. The novel everolimus-eluting, platinum chromium SYNERGY stent is coated with a bioabsorbable abluminal polymer that resolves within 4 months. METHODS This was a prospective, single center registry of consecutive patients treated with the SYNERGY stent between December 2012 and April 2015. The primary outcome was the incidence of TLF, defined as the combination of cardiac death, target vessel myocardial infarction, or clinically driven target lesion revascularization (TLR) at 1 year. RESULTS A total of 807 patients received the SYNERGY stent during the study period. One-year clinical outcome data was available for 765 patients (94.8%) and were considered for statistical analysis. The mean age was 60.7 ± 10.8 years, and 83.4% were males. Patients with acute myocardial infarction consisted of 50.3% (ST-segment elevation myocardial infarction: 23.0%, Non-ST-segment elevation myocardial infarction: 27.3%) of the study population. The treated lesions were complex (ACC/AHA type B2/C: 72.7%). The primary end point of TLF at 1 year was 5.8%. Rates of cardiac mortality, target vessel myocardial infarction, and TLR were 4.2, 1.0, and 1.3%, respectively, at 1 year. Predictors of the incidence and time to early TLF were female gender, Malay ethnicity, diabetes mellitus, acute myocardial infarction at presentation, a prior history of coronary artery bypass surgery and the presence of lesion calcification. The incidence of definite stent thrombosis was 0.4% at 1 year. CONCLUSIONS In this registry, the use of the SYNERGY stent was associated with low rates of TLF at 1 year.
Collapse
Affiliation(s)
| | - William Kristanto
- Department of Cardiology, National University Heart Centre, Singapore
| | - Li Liu
- Department of Cardiology, National University Heart Centre, Singapore
| | - Siew-Pang Chan
- Department of Cardiology, National University Heart Centre, Singapore
| | - Poay Huan Loh
- Department of Cardiology, National University Heart Centre, Singapore
| | - Edgar L Tay
- Department of Cardiology, National University Heart Centre, Singapore
| | - Koo Hui Chan
- Department of Cardiology, National University Heart Centre, Singapore
| | - Mark Y Chan
- Department of Cardiology, National University Heart Centre, Singapore
| | - Chi-Hang Lee
- Department of Cardiology, National University Heart Centre, Singapore
| | - Adrian F Low
- Department of Cardiology, National University Heart Centre, Singapore
| | - Huay Cheem Tan
- Department of Cardiology, National University Heart Centre, Singapore
| | - Joshua P Loh
- Department of Cardiology, National University Heart Centre, Singapore
| |
Collapse
|
905
|
Puurunen MK, Hwang SJ, Larson MG, Vasan RS, O'Donnell CJ, Tofler G, Johnson AD. ADP Platelet Hyperreactivity Predicts Cardiovascular Disease in the FHS (Framingham Heart Study). J Am Heart Assoc 2018; 7:JAHA.118.008522. [PMID: 29502103 PMCID: PMC5866343 DOI: 10.1161/jaha.118.008522] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Platelet function is associated with adverse events in patients with cardiovascular disease (CVD). METHODS AND RESULTS We examined associations of baseline platelet function with incident CVD events in the community-based FHS (Framingham Heart Study). Participants free of prevalent CVD and without recent aspirin treatment with available data in the Framingham Offspring cohort (1991-1995) and Omni cohort (1994-1998) were included. Platelet function was measured with light transmission aggregometry using collagen (1.9 μg/mL), ADP (0.05-15 μmol/L), and epinephrine (0.01-15 μmol/L). We used proportional hazards models to analyze incident outcomes (myocardial infarction/stroke, CVD, and CVD mortality) with respect to platelet measures. The study sample included 2831 participants (average age, 54.3 years; 57% women). During follow-up (median, 20.4 years), we observed 191 composite incident myocardial infarction or stroke events, 432 incident CVD cases, and 117 CVD deaths. Hyperreactivity to ADP and platelet aggregation at ADP concentration of 1.0 μmol/L were significantly associated with incident myocardial infarction/stroke in a multivariable model (hazard ratio, 1.68 [95% confidence interval, 1.13-2.50] [P=0.011] for hyperreactivity across ADP doses; and hazard ratio, 1.16 [95% confidence interval, 1.02-1.33] [P=0.029] for highest quartile of ADP response at 1.0 μmol/L versus others). No association was observed for collagen lag time or any epinephrine measures with incident myocardial infarction or stroke. CONCLUSIONS Intrinsic hyperreactivity to low-dose ADP in our community-based sample, who were free of CVD and any antiplatelet therapy, is associated with future arterial thrombosis during a 20-year follow-up. These findings reinforce ADP activation inhibition as a critical treatment paradigm and encourage further study of ADP inhibitor-refractive populations.
Collapse
Affiliation(s)
- Marja K Puurunen
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Schools of Medicine and Public Health, Boston University, Boston, MA
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Framingham, MA
| | - Martin G Larson
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Biostatistics Department, Boston University School of Public Health, Boston, MA
| | - Ramachandran S Vasan
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Schools of Medicine and Public Health, Boston University, Boston, MA
| | - Christopher J O'Donnell
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Framingham, MA
| | - Geoffrey Tofler
- Royal North Shore Hospital, Sydney, New South Wales, Australia.,University of Sydney, New South Wales, Australia
| | - Andrew D Johnson
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA .,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Framingham, MA
| |
Collapse
|
906
|
Abu-Assi E, Raposeiras-Roubin S, Cobas-Paz R, Caneiro-Queija B, Martínez-Reglero C, Rodríguez-Rodríguez JM, Baz A, Íñiguez-Romo A. Assessing the performance of the PRECISE-DAPT and PARIS risk scores for predicting one-year out-of-hospital bleeding in acute coronary syndrome patients. EUROINTERVENTION 2018; 13:1914-1922. [DOI: 10.4244/eij-d-17-00550] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
907
|
Parviz Y, Shlofmitz E, Fall KN, Konigstein M, Maehara A, Jeremias A, Shlofmitz RA, Mintz GS, Ali ZA. Utility of intracoronary imaging in the cardiac catheterization laboratory: comprehensive evaluation with intravascular ultrasound and optical coherence tomography. Br Med Bull 2018; 125:79-90. [PMID: 29360941 DOI: 10.1093/bmb/ldx049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/15/2018] [Indexed: 11/14/2022]
Abstract
BACKGROUND Intracoronary imaging is an important tool for guiding decision making in the cardiac catheterization laboratory. SOURCES OF DATA We have reviewed the latest available evidence in the field to highlight the various potential benefits of intravascular imaging. AREAS OF AGREEMENT Coronary angiography has been considered the gold standard test to appropriately diagnose and manage patients with coronary artery disease, but it has the inherent limitation of being a 2-dimensional x-ray lumenogram of a complex 3-dimensional vascular structure. AREAS OF CONTROVERSY There is well-established inter- and intra-observer variability in reporting coronary angiograms leading to potential variability in various management strategies. Intracoronary imaging improves the diagnostic accuracy while optimizing the results of an intervention. Utilization of intracoronary imaging modalities in routine practice however remains low worldwide. Increased costs, resources, time and expertise have been cited as explanations for low incorporation of these techniques. GROWING POINTS Intracoronary imaging supplements and enhances an operator's decision-making ability based on detailed and objective lesion assessment rather than a subjective visual estimation. The benefits of intravascular imaging are becoming more profound as the complexity of cases suitable for revascularization increases. AREAS TIMELY FOR DEVELOPING RESEARCH While the clinical benefits of intravascular ultrasound have been well validated, optical coherence tomography in comparison is a newer technology, with robust clinical trials assessing its clinical benefit are underway.
Collapse
Affiliation(s)
- Yasir Parviz
- Division of Cardiology, Columbia UniversityMedical Center, New York, NY, USA
| | - Evan Shlofmitz
- Division of Cardiology, Columbia University Medical Center, New York, NY, USA.,Cardiovascular Research Foundation, New York, NY, USA
| | - Khady N Fall
- Division of Cardiology, Columbia UniversityMedical Center, New York, NY, USA
| | | | - Akiko Maehara
- Division of Cardiology, Columbia University Medical Center, New York, NY, USA.,Cardiovascular Research Foundation, New York, NY, USA
| | - Allen Jeremias
- Cardiovascular Research Foundation, New York, NY, USA.,St. Francis Hospital, Roslyn, NY, USA
| | | | - Gary S Mintz
- Cardiovascular Research Foundation, New York, NY, USA
| | - Ziad A Ali
- Division of Cardiology, Columbia University Medical Center, New York, NY, USA.,Cardiovascular Research Foundation, New York, NY, USA
| |
Collapse
|
908
|
Horne BD, Muhlestein JB, Bhandary D, Hoetzer GL, Khan ND, Bair TL, Lappé DL. Clinically feasible stratification of 1-year to 3-year post-myocardial infarction risk. Open Heart 2018. [PMID: 29531761 PMCID: PMC5845421 DOI: 10.1136/openhrt-2017-000723] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective Post-myocardial infarction (MI) care is crucial to preventing recurrent major adverse cardiovascular events (MACE), but can be complicated to personalise. A tool is needed that effectively stratifies risk of cardiovascular (CV) events 1–3 years after MI but is also clinically usable. Methods Patients surviving ≥1 year after an index MI with ≥1 risk factor for recurrent MI (ie, age ≥65 years, prior MI, multivessel coronary disease, diabetes, glomerular filtration rate <60 mL/min/1.73 m2) were studied. Cox regression derived sex-specific Intermountain Major Adverse Cardiovascular Events (IMACE) risk scores for the composite of 1-year to 3-year MACE (CV death, MI or stroke). Derivation was performed in 70% of subjects (n=1342 women; 3047 men), with validation in the other 30% (n=576 women; 1290 men). Secondary validations were also performed. Results In women, predictors of CV events were glucose, creatinine, haemoglobin, platelet count, red cell distribution width (RDW), age and B-type natriuretic peptide (BNP); among men, they were potassium, glucose, blood urea nitrogen, haematocrit, white blood cell count, RDW, mean platelet volume, age and BNP. In the primary validation, in women, IMACE ranged from 0 to 11 (maximum possible: 12) and had HR=1.44 per +1 score (95% CI 1.29 to 1.61; P<0.001); men had IMACE range 0–14 (maximum: 16) and HR=1.29 per +1 score (95% CI 1.20 to 1.38; P<0.001). IMACE ≥5 in women (≥6 in men) showed strikingly higher MACE risk. Conclusions Sex-specific risk scores strongly stratified 1-year to 3-year post-MI MACE risk. IMACE is an inexpensive, dynamic, electronically delivered tool for evaluating and better managing post-MI patient care.
Collapse
Affiliation(s)
- Benjamin D Horne
- Intermountain Heart Institute, Intermountain Medical Center, Salt Lake City, Utah, USA.,Department of Biomedical Informatics, University of Utah, Salt Lake City, Utah, USA
| | - Joseph B Muhlestein
- Intermountain Heart Institute, Intermountain Medical Center, Salt Lake City, Utah, USA.,Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | | | | | - Naeem D Khan
- AstraZeneca Pharmaceuticals LP, Wilmington, Delaware, USA
| | - Tami L Bair
- Intermountain Heart Institute, Intermountain Medical Center, Salt Lake City, Utah, USA
| | - Donald L Lappé
- Intermountain Heart Institute, Intermountain Medical Center, Salt Lake City, Utah, USA.,Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
909
|
Harima A, Sairaku A, Inoue I, Nishioka K, Oka T, Nakama Y, Dai K, Ohi K, Hashimoto H, Kihara Y. Real-life experience of a stent-less revascularization strategy using a combination of excimer laser and drug-coated balloon for patients with acute coronary syndrome. J Interv Cardiol 2018; 31:284-292. [PMID: 29464846 DOI: 10.1111/joic.12495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/09/2018] [Accepted: 01/18/2018] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES We aimed to test a novel stent-less revascularization strategy using a combination of excimer laser coronary angioplasty (ELCA) and drug-coated balloon (DCB) for patients with acute coronary syndrome (ACS). BACKGROUND Percutaneous coronary intervention with drug eluting stents is a standard invasive treatment for ACS. Some unsolved issues however remain, such as stent thrombosis and bleeding risks associated with dual antiplatelet therapy. METHODS Consecutive ACS patients were planned to receive either a DCB application following ELCA without a stent implantation or conventional revascularization with a coronary stent. The endpoints were (i) major cardiac adverse events (MACEs), defined as the composite of cardiac death, myocardial infarctions, and target lesion revascularization; (ii) target vessel revascularization (TVR); and (iii) angiographic outcome. RESULTS Since a greater than expected number of patients allocated to the stent-less treatment arm eventually received a bailout stenting, the following 3 as-treated groups were compared; DCB with ELCA group (N = 60), Stent with ELCA group (N = 23), and Stent without ELCA group (N = 85). During a mean follow-up period of 420 ± 137 days, and with angiographic 6- and 12-month-follow-up rates of 96.7%, 87%, and 81.2%, and 50%, 65.2%, and 45.9%, respectively, the MACE rate did not differ across the groups (10%, 4.3%, and 3.5%; P = 0.22) while an incidence of TVR was more common (15%, 0, and 4.7%; P = 0.02) and the diameter stenosis at 6-months of follow-up was greater (25.7 ± 18.2, 14.9 ± 13.1 and 16.2 ± 15.4%; P = 0.002) in the DCB with ELCA group. CONCLUSIONS The stent-less revascularization strategy with DCB and ELCA was associated with a higher occurrence of restenosis in ACS patients.
Collapse
Affiliation(s)
- Ayako Harima
- Department of Cardiology, Hiroshima City Hospital, Hiroshima, Japan
| | - Akinori Sairaku
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Ichiro Inoue
- Department of Cardiology, Hiroshima City Hospital, Hiroshima, Japan
| | - Kenji Nishioka
- Department of Cardiology, Hiroshima City Hospital, Hiroshima, Japan
| | - Toshiharu Oka
- Department of Cardiology, Hiroshima City Hospital, Hiroshima, Japan
| | - Yasuharu Nakama
- Department of Cardiology, Hiroshima City Hospital, Hiroshima, Japan
| | - Kazuoki Dai
- Department of Cardiology, Hiroshima City Hospital, Hiroshima, Japan
| | - Kuniomi Ohi
- Department of Cardiology, Hiroshima City Hospital, Hiroshima, Japan
| | - Haruki Hashimoto
- Department of Cardiology, Hiroshima City Hospital, Hiroshima, Japan.,Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
910
|
Yoshikawa Y, Shiomi H, Watanabe H, Natsuaki M, Kondo H, Tamura T, Nakagawa Y, Morimoto T, Kimura T. Validating Utility of Dual Antiplatelet Therapy Score in a Large Pooled Cohort From 3 Japanese Percutaneous Coronary Intervention Studies. Circulation 2018; 137:551-562. [DOI: 10.1161/circulationaha.117.028924] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/18/2017] [Indexed: 11/16/2022]
Abstract
Background:
The dual antiplatelet therapy (DAPT) score was developed to estimate ischemic and bleeding risks from the DAPT study. However, few studies validated its utility externally. We sought to validate the utility of the DAPT score in the Japanese population.
Methods:
In a pooled cohort of 3 studies conducted in Japan (the CREDO-Kyoto [Coronary Revascularization Demonstrating Outcome Study in Kyoto] Registry Cohort-2, RESET [Randomized Evaluation of Sirolimus-Eluting Versus Everolimus-Eluting Stent Trial], and NEXT [NOBORI Biolimus-Eluting Versus XIENCE/PROMUS Everolimus-Eluting Stent Trial]), we compared risks for ischemic and bleeding events from 13 to 36 months after percutaneous coronary intervention among patients with a DAPT score ≥2 (high DS) and a DAPT score <2 (low DS).
Results:
Among 12 223 patients receiving drug-eluting stents who were free from ischemic or bleeding events at 13 months after percutaneous coronary intervention, 3944 patients had high DS and 8279 had low DS. The cumulative incidence of primary ischemic end point (myocardial infarction/stent thrombosis) was significantly higher in high DS than in low DS (1.5% versus 0.9%,
P
=0.002), whereas the cumulative incidence of primary bleeding end point (GUSTO moderate/severe) tended to be lower in high DS than in low DS (2.1% versus 2.7%,
P
=0.07). The cumulative incidences of cardiac death, myocardial infarction, and stent thrombosis were also significantly higher in high DS than in low DS (2.0% versus 1.4%,
P
=0.03; 1.5% versus 0.8%,
P
=0.002; 0.7% versus 0.3%,
P
<0.001, respectively), whereas the cumulative incidences of noncardiac death and GUSTO severe bleeding were significantly lower in high DS than in low DS (2.4% versus 3.9%,
P
<0.001; 1.0% versus 1.6%,
P
=0.03, respectively).
Conclusions:
In the current population, the DAPT score successfully stratified ischemic and bleeding risks, although the ischemic event rate was remarkably low even in high DS. Further studies would be warranted to evaluate the utility of prolonged DAPT guided by the DAPT score.
Collapse
Affiliation(s)
- Yusuke Yoshikawa
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Japan (Y.Y., H.S., H.W., T.K.)
| | - Hiroki Shiomi
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Japan (Y.Y., H.S., H.W., T.K.)
| | - Hirotoshi Watanabe
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Japan (Y.Y., H.S., H.W., T.K.)
| | - Masahiro Natsuaki
- Department of Cardiovascular Medicine, Saga University, Japan (M.N.)
| | - Hirokazu Kondo
- Department of Cardiology, Tenri Hospital, Japan (H.K., T.T., Y.N.)
| | - Toshihiro Tamura
- Department of Cardiology, Tenri Hospital, Japan (H.K., T.T., Y.N.)
| | | | - Takeshi Morimoto
- Department of Clinical Epidemiology, Hyogo College of Medicine, Nishinomiya, Japan (T.M.)
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Japan (Y.Y., H.S., H.W., T.K.)
| |
Collapse
|
911
|
Singh M, Raghavan D, Williams JS, Martin BC, Hudson TJ, Owen RR, Jain N. Prevalence of Chronic Kidney Disease, Thrombotic Cardiovascular Events, and Use of Oral P2Y12 Inhibitors among Veterans. Am J Nephrol 2018; 47:67-71. [PMID: 29393120 DOI: 10.1159/000486647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/06/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Contemporary prevalence of chronic kidney disease (CKD) and thrombotic cardiovascular (CV) events remains unclear in Veterans enrolled in the Veterans Affairs Health Care System (VA) care. Although oral P2Y12 inhibitors (P2Y12i) are increasingly being prescribed to this patient population, the overall prescription trend for P2Y12i remains unclear. METHODS Using national VA corporate warehouse data, we used International Classification of Diseases-9 codes to identify Veterans with CKD, dialysis-dependent CKD, and CV events. VA pharmacy data were used to count P2Y12i prescriptions for the federal fiscal years (FY) 2011 through 2015. RESULTS The period prevalence of Veterans with CKD was 378,233 (6.1%). The point prevalence of CKD increased by 49% from 132,979 (2.30%) in FY11 to 213,444 (3.42%) in FY15. The period prevalence of Veterans with dialysis-dependent CKD was 150,298 (2.4%). In all, 128,703 (56.7%) CV events occurred in Veterans with CKD. Veterans with CKD were given 50.1% of prescriptions for clopidogrel, 49.3% for prasugrel, and 60.4% for ticagrelor. In this patient population, year-to-year increases in P2Y12i prescriptions were observed with a dramatic increase in ticagrelor prescriptions. CONCLUSION CKD is common among Veterans and its true prevalence is likely being underestimated. The prevalence of dialysis-dependent CKD is higher among Veterans than the non-Veteran US population. CV events are widely co-prevalent and these patients are commonly prescribed P2Y12i. The recent increase in ticagrelor prescriptions in this patient population and large cost differences between the 3 P2Y12i underline the need for future studies to identify the preferred P2Y12i for these patients.
Collapse
Affiliation(s)
- Manisha Singh
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Central Arkansas Veterans Affairs Healthcare System, Little Rock, Arkansas, USA
| | - Deepa Raghavan
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Central Arkansas Veterans Affairs Healthcare System, Little Rock, Arkansas, USA
| | - James S Williams
- Central Arkansas Veterans Affairs Healthcare System, Little Rock, Arkansas, USA
| | - Bradley C Martin
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Teresa J Hudson
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Central Arkansas Veterans Affairs Healthcare System, Little Rock, Arkansas, USA
| | - Richard R Owen
- Central Arkansas Veterans Affairs Healthcare System, Little Rock, Arkansas, USA
| | - Nishank Jain
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Central Arkansas Veterans Affairs Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
912
|
Mori M, Shioda K, Yun JJ, Mangi AA, Darr U, Geirsson A. Pattern and predictors of dual antiplatelet use after coronary artery bypass graft surgery. J Thorac Cardiovasc Surg 2018; 155:632-638. [DOI: 10.1016/j.jtcvs.2017.09.092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 08/28/2017] [Accepted: 09/19/2017] [Indexed: 11/26/2022]
|
913
|
|
914
|
Spinthakis N, Farag M, Rocca B, Gorog DA. More, More, More: Reducing Thrombosis in Acute Coronary Syndromes Beyond Dual Antiplatelet Therapy-Current Data and Future Directions. J Am Heart Assoc 2018; 7:e007754. [PMID: 29374045 PMCID: PMC5850258 DOI: 10.1161/jaha.117.007754] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | - Mohamed Farag
- Postgraduate Medical School, University of Hertfordshire, United Kingdom
| | - Bianca Rocca
- Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - Diana A Gorog
- Postgraduate Medical School, University of Hertfordshire, United Kingdom
- National Heart & Lung Institute Imperial College, London, United Kingdom
| |
Collapse
|
915
|
Xu JJ, Gao Z, Zhang Y, Gao LJ, Chen J, Qiao SB, Gao RL, Yang YJ, Xu B, Yuan JQ. Dual antiplatelet therapy after coronary drug-eluting stent implantation in China: A large single center study. Catheter Cardiovasc Interv 2018; 91:566-572. [PMID: 29359390 DOI: 10.1002/ccd.27500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/27/2017] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To investigate the actual dual antiplatelet therapy (DAPT) duration after drug-eluting stent (DES) treatment in China. BACKGROUND Currently, less is known about actual DAPT duration after drug-eluting stent (DES) treatment in China. Here, we performed a study in the largest cardiovascular center in China to investigate DAPT duration and identify associated factors after DES implantation. METHODS A total of 9,919 consecutive patients with DES implantation from January 2013 to December 2013 were enrolled. DAPT cessation was observed, and factors associated with different DAPT durations were analyzed. RESULTS The median follow-up time was 882 days. The proportion of patients with DAPT coverage at 1-year of follow-up was 97.3%, and it decreased to 30.1% for 2 years. The distribution of DAPT duration was not significantly different among patients with acute myocardial infarction (AMI) versus non-AMI (P = 0.41) and with new-generation DES versus first-generation DES (P = 0.54). The multivariable analysis indicated some independent predictors prolonging DAPT duration, including target vessel revascularization (OR 2.50, 95% CI 2.04-3.06, P < 0.001), stent numbers (OR 1.10, 95% CI 1.05-1.15, P < 0.001), and previous coronary artery bypass grafting (OR 0.76, 95% CI 0.61-0.96, P = 0.02). Other clinical factors, such as the increased risk of bleeding and high ischemic risk, were not associated with DAPT duration. CONCLUSIONS The 1-year DAPT after DES was applied to 97.3% of Chinese patients in the studied clinical center. However, the DAPT duration after 1 year was not adjusted according to the patients' bleeding situation and ischemic risks.
Collapse
Affiliation(s)
- Jing-Jing Xu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhan Gao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yin Zhang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Li-Jian Gao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jue Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Shu-Bin Qiao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Run-Lin Gao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yue-Jin Yang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Bo Xu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jin-Qing Yuan
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
916
|
Chen ST, Patel MR. Comparison of Anticoagulant Therapy for Atrial Fibrillation - Novel Oral Anticoagulants Versus Vitamin K Antagonists. Prog Cardiovasc Dis 2018; 60:514-523. [PMID: 29339167 DOI: 10.1016/j.pcad.2018.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/11/2018] [Indexed: 12/15/2022]
Abstract
In patients with non-valvular atrial fibrillation (NVAF), oral anticoagulation is important for prevention of stroke and systemic embolism (SE). While Vitamin K antagonists (VKAs) have historically been the standard of care, these medications are limited by numerous food and drug interactions with onerous requirements for frequent monitoring and dose adjustments. Over the past decade, several novel oral anticoagulants (NOACs) have been developed to directly inhibit factor IIa/thrombin (dabigatran) or activated factor X (apixaban, rivaroxaban, edoxaban). These medications have been shown to be at least as effective as warfarin for stroke prevention in NVAF with more favorable safety profiles. However, their advantages are underscored by a lack of specific antidotes and assays quantifying their anticoagulant effects. This paper addresses the use of NOACs compared to VKAs in patients with NVAF, with a special focus on high-risk populations, including the elderly, those with renal disease, diabetes mellitus, coronary artery disease, and previous stroke. The current literature surrounding special clinical scenarios including the treatment of bleeding, perioperative management, and the use of NOACs in cardioversion and catheter ablation will be also discussed.
Collapse
Affiliation(s)
- Sean T Chen
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, United States
| | - Manesh R Patel
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
917
|
Tantry US, Navarese EP, Myat A, Gurbel PA. Selection of P2Y 12 Inhibitor in Percutaneous Coronary Intervention and/or Acute Coronary Syndrome. Prog Cardiovasc Dis 2018; 60:460-470. [PMID: 29339168 DOI: 10.1016/j.pcad.2018.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 01/10/2018] [Indexed: 01/02/2023]
Abstract
The P2Y12 receptor plays a critical role in the amplification of platelet aggregation in response to various agonists and stable thrombus generation at the site of vascular injury leading to deleterious ischemic complications. Therefore, treatment with a P2Y12 receptor blocker is a major effective strategy to prevent ischemic complications in high-risk patients with acute coronary syndrome (ACS) and patients undergoing percutaneous coronary intervention (PCI). The determination of optimal platelet inhibition is based on maximizing antithrombotic properties while minimizing bleeding risk and is critically dependent on individual patient's propensity for thrombotic and bleeding risks. Immediately after ACS and during PCI, where highly elevated thrombotic activity is present, a loading dose administration with a potent P2Y12 receptor blocker such as ticagrelor or prasugrel is preferred. In stable coronary artery disease patients undergoing PCI, clopidogrel is widely used. In addition, in patients with ST-segment elevation myocardial infraction who cannot take oral medications, a fast acting intravenous glycoprotein IIb/IIIa inhibitor or P2Y12 receptor blocker, cangrelor, may add clinical benefits. During long term therapy, a strategy that prevents ischemic risk while avoiding excessive bleeding risk is similarly desired. Although up to one year dual antiplatelet therapy (DAPT) is recommended in patients undergoing elective stenting, the available data support the anti-ischemic benefit of prolonged DAPT (more than1 year) in patients with prior MI. In addition to the DAPT risk calculator tool, future risk assessment methods that analyze intrinsic thrombogenicity and atherosclerotic coronary burden may further identify the optimal candidate for prolonged DAPT to improve net clinical outcomes.
Collapse
Affiliation(s)
- Udaya S Tantry
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Falls Church, VA, USA
| | - Eliano P Navarese
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Falls Church, VA, USA
| | - Aung Myat
- Sussex Cardiac Centre, Brighton and Sussex University Hospitals NHS Trust and Faculty of Medicine, Brighton and Sussex Medical School, Brighton, UK
| | - Paul A Gurbel
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Falls Church, VA, USA.
| |
Collapse
|
918
|
Seara FAC, Maciel L, Barbosa RAQ, Rodrigues NC, Silveira ALB, Marassi MP, Carvalho AB, Nascimento JHM, Olivares EL. Cardiac ischemia/reperfusion injury is inversely affected by thyroid hormones excess or deficiency in male Wistar rats. PLoS One 2018; 13:e0190355. [PMID: 29304184 PMCID: PMC5755761 DOI: 10.1371/journal.pone.0190355] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/13/2017] [Indexed: 11/19/2022] Open
Abstract
AIM Thyroid dysfunctions can increase the risk of myocardial ischemia and infarction. However, the repercussions on cardiac ischemia/reperfusion (IR) injury remain unclear so far. We report here the effects of hypothyroidism and thyrotoxicosis in the susceptibility to IR injury in isolated rat hearts compared to euthyroid condition and the potential role of antioxidant enzymes. METHODS Hypothyroidism and thyrotoxicosis were induced by administration of methimazole (MMZ, 300 mg/L) and thyroxine (T4, 12 mg/L), respectively in drinking water for 35 days. Isolated hearts were submitted to IR and evaluated for mechanical dysfunctions and infarct size. Superoxide dismutase types 1 and 2 (SOD1 and SOD2), glutathione peroxidase types 1 and 3 (GPX 1 and GPX3) and catalase mRNA levels were assessed by quantitative RT-PCR to investigate the potential role of antioxidant enzymes. RESULTS Thyrotoxicosis elicited cardiac hypertrophy and increased baseline mechanical performance, including increased left ventricle (LV) systolic pressure, LV developed pressure and derivatives of pressure (dP/dt), whereas in hypothyroid hearts exhibited decreased dP/dt. Post-ischemic recovery of LV end-diastolic pressure (LVEDP), LVDP and dP/dt was impaired in thyrotoxic rat hearts, whereas hypothyroid hearts exhibited improved LVEDP and decreased infarct size. Catalase expression was decreased by thyrotoxicosis. CONCLUSION Thyrotoxicosis was correlated, at least in part, to cardiac remodeling and increased susceptibility to IR injury possibly due to down-regulation of antioxidant enzymes, whereas hypothyroid hearts were less vulnerable to IR injury.
Collapse
Affiliation(s)
- Fernando A. C. Seara
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Institute of Biology, Federal Rural University of Rio de Janeiro, Seropedica–RJ, Brazil
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro–Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Maciel
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro–Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raiana A. Q. Barbosa
- Laboratory of Cellular and Molecular Cardiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro–Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nayana C. Rodrigues
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Institute of Biology, Federal Rural University of Rio de Janeiro, Seropedica–RJ, Brazil
| | - Anderson L. B. Silveira
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Institute of Biology, Federal Rural University of Rio de Janeiro, Seropedica–RJ, Brazil
- Laboratory of Physiology and Human Performance, Department of Physical Education and Sports, Institute of Education, Federal Rural University of Rio de Janeiro, Seropedica–RJ, Brazil
| | - Michelle P. Marassi
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Institute of Biology, Federal Rural University of Rio de Janeiro, Seropedica–RJ, Brazil
| | - Adriana B. Carvalho
- Laboratory of Cellular and Molecular Cardiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro–Rio de Janeiro, Rio de Janeiro, Brazil
| | - José Hamilton M. Nascimento
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro–Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emerson L. Olivares
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Institute of Biology, Federal Rural University of Rio de Janeiro, Seropedica–RJ, Brazil
| |
Collapse
|
919
|
Sellers D, Srinivas C, Djaiani G. Cardiovascular complications after non-cardiac surgery. Anaesthesia 2018; 73 Suppl 1:34-42. [DOI: 10.1111/anae.14138] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2017] [Indexed: 01/04/2023]
Affiliation(s)
- D. Sellers
- Toronto General Hospital; University of Toronto; Toronto Canada
| | - C. Srinivas
- Toronto General Hospital; University of Toronto; Toronto Canada
| | - G. Djaiani
- Toronto General Hospital; University of Toronto; Toronto Canada
| |
Collapse
|
920
|
Abstract
Antiplatelet drugs, such as aspirin, P2Y12 antagonists, and glycoprotein (GP) IIb/IIIa inhibitors, have proved to be successful in reducing the morbidity and mortality associated with arterial thrombosis. These agents are, therefore, the cornerstone of therapy for patients with acute coronary syndromes. However, these drugs all carry an inherent risk of bleeding, which is associated with adverse cardiovascular outcomes and mortality. Thus, the potential benefits of more potent, conventional antiplatelet drugs are likely be offset by the increased risk of bleeding. Data from experiments in vivo have highlighted potentially important differences between haemostasis and thrombosis, raising the prospect of developing new antiplatelet drugs that are not associated with bleeding. Indeed, in preclinical studies, several novel antiplatelet therapies that seem to inhibit thrombosis while maintaining haemostasis have been identified. These agents include inhibitors of phosphatidylinositol 3-kinase-β (PI3Kβ), protein disulfide-isomerase, activated GPIIb/IIIa, GPIIb/IIIa outside-in signalling, protease-activated receptors, and platelet GPVI-mediated adhesion pathways. In this Review, we discuss how a therapeutic ceiling has been reached with existing antiplatelet drugs, whereby increased potency is offset by elevated bleeding risk. The latest advances in our understanding of thrombus formation have informed the development of new antiplatelet drugs that are potentially safer than currently available therapies.
Collapse
|
921
|
Kerneis M, Talib U, Nafee T, Daaboul Y, Pahlavani S, Pitliya A, Furqan M, Datta S, Kazmi HA, Younes A, Gibson CM. Triple Antithrombotic Therapy for Patients with Atrial Fibrillation Undergoing Percutaneous Coronary Intervention. Prog Cardiovasc Dis 2018; 60:524-530. [DOI: 10.1016/j.pcad.2018.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 01/17/2018] [Indexed: 12/11/2022]
|
922
|
The Society for Vascular Surgery practice guidelines on the care of patients with an abdominal aortic aneurysm. J Vasc Surg 2018; 67:2-77.e2. [DOI: 10.1016/j.jvs.2017.10.044] [Citation(s) in RCA: 1150] [Impact Index Per Article: 164.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
923
|
Comparison of prasugrel versus clopidogrel in Korean patients with acute myocardial infarction undergoing successful revascularization. J Cardiol 2018; 71:36-43. [DOI: 10.1016/j.jjcc.2017.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/01/2017] [Accepted: 05/22/2017] [Indexed: 11/21/2022]
|
924
|
Agarwal N, Mahmoud AN, Patel NK, Jain A, Garg J, Mojadidi MK, Agrawal S, Qamar A, Golwala H, Gupta T, Bhatia N, Anderson RD, Bhatt DL. Meta-Analysis of Aspirin Versus Dual Antiplatelet Therapy Following Coronary Artery Bypass Grafting. Am J Cardiol 2018; 121:32-40. [PMID: 29122278 DOI: 10.1016/j.amjcard.2017.09.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 11/20/2022]
Abstract
Although aspirin monotherapy is considered the standard of care after coronary artery bypass grafting (CABG), more recent evidence has suggested a benefit with dual antiplatelet therapy (DAPT) after CABG. We performed a meta-analysis of observational studies and randomized controlled trials comparing outcomes of aspirin monotherapy with DAPT in patients after CABG. Subgroup analyses were conducted according to surgical technique (i.e., on vs off pump) and clinical presentation (acute coronary syndrome vs no acute coronary syndrome). Random effects overall risk ratios (RR) were calculated using the DerSimonian and Laird model. Eight randomized control trials and 9 observational studies with a total of 11,135 patients were included. At a mean follow-up of 23 months, major adverse cardiac events (10.3% vs 12.1%, RR 0.84, confidence interval [CI] 0.71 to 0.99), all-cause mortality (5.7% vs 7.0%, RR 0.67, CI 0.48 to 0.94), and graft occlusion (11.3% vs 14.2%, RR 0.79, CI 0.63 to 0.98) were less with DAPT than with aspirin monotherapy. There was no difference in myocardial infarction, stroke, or major bleeding between the 2 groups. In conclusion, DAPT appears to be associated with a reduction in graft occlusion, major adverse cardiac events, and all-cause mortality, without significantly increasing major bleeding compared with aspirin monotherapy in patients undergoing CABG.
Collapse
Affiliation(s)
- Nayan Agarwal
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Ahmed N Mahmoud
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Nimesh Kirit Patel
- Department of Medicine, Virginia Commonwealth University Health System, Richmond, Virginia
| | - Ankur Jain
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Jalaj Garg
- Department of Medicine, Lehigh Valley, Allentown, Pennsylvania
| | | | - Sahil Agrawal
- Department of Medicine, St Lukes University Health Network, Bethlehem, Pennsylvania
| | - Arman Qamar
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Harsh Golwala
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Tanush Gupta
- Department of Medicine, Montefiore Medical Centre, Albert Einstein College of Medicine, Bronx, New York
| | - Nirmanmoh Bhatia
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - R David Anderson
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Deepak L Bhatt
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| |
Collapse
|
925
|
Kang J, Kim HS. The Evolving Concept of Dual Antiplatelet Therapy after Percutaneous Coronary Intervention: Focus on Unique Feature of East Asian and "Asian Paradox". Korean Circ J 2018; 48:537-551. [PMID: 29968428 PMCID: PMC6031716 DOI: 10.4070/kcj.2018.0166] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/05/2018] [Indexed: 12/20/2022] Open
Abstract
Dual antiplatelet therapy (DAPT) with aspirin and a P2Y12 inhibitor is essential after percutaneous coronary intervention (PCI), while many studies have focused on determining the optimal degree of platelet inhibition and optimal DAPT duration to minimize complications after PCI. Current guidelines developed by the American College of Cardiology/American Heart Association and the European Society of Cardiology summarize previous studies and provide recommendations. However, these guidelines are mainly based on Western patients, and their characteristics might differ from those of East Asian patients. Previous data suggested that East Asian patients have unique features with regard to the response to antiplatelet agents. On comparing Western and East Asian patients, it was found that East Asian patients have a lower rate of ischemic events and higher rate of bleeding events after PCI, despite a higher on-treatment platelet reactivity, which is referred to as the “East Asian paradox.” As the main purpose of DAPT is to minimize ischemic and bleeding complications after PCI, these differences should be clarified before adopting the guidelines for East Asian patients. Therefore, in this article, we will review various issues regarding DAPT in East Asian patients, with a focus on the unique characteristics of East Asian patients, previous studies regarding antiplatelet agents in East Asian patients, and a guideline from an East Asian perspective.
Collapse
Affiliation(s)
- Jeehoon Kang
- Department of Internal Medicine, Cardiovascular Center, Seoul National University Hospital, Seoul, Korea
| | - Hyo Soo Kim
- Department of Internal Medicine, Cardiovascular Center, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
926
|
Dugani S, Wasan KM, Kissoon N. World Health Organization and Essential Medicines. J Pharm Sci 2017; 107:1261-1262. [PMID: 29277641 DOI: 10.1016/j.xphs.2017.12.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/14/2017] [Accepted: 12/18/2017] [Indexed: 10/18/2022]
Abstract
In June 2017, the World Health Organization released 20th Model List of Essential Medicines for adults and sixth Model List of Essential Medicines for children. In our commentary, we describe the changes to the Essential Medicine list, and identify deficits in excluding medicines for management of diseases with a high burden. In using tracer conditions such as cardiovascular and thromboembolic disease, mental health, and diseases of the musculoskeletal system, we highlight the absence of several medicines, which are incorporated into major clinical practice guidelines. We recommend that the World Health Organization review its process with respect to identifying disease conditions as well as evidence-based therapies.
Collapse
Affiliation(s)
- Sagar Dugani
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, Minnesota 55902.
| | - Kishor M Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Niranjan Kissoon
- University of British Columbia and British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
927
|
Disseminated Mycobacterium chimaera infection associated with heater-cooler units after aortic valve surgery without endocarditis. J Thorac Cardiovasc Surg 2017; 155:2369-2374. [PMID: 29397149 DOI: 10.1016/j.jtcvs.2017.12.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/16/2017] [Accepted: 12/03/2017] [Indexed: 12/17/2022]
|
928
|
Zhu P, Gao Z, Tang XF, Xu JJ, Zhang Y, Gao LJ, Chen J, Qiao SB, Yang YJ, Gao RL, Xu B, Yuan JQ. Impact of Proton-pump Inhibitors on the Pharmacodynamic Effect and Clinical Outcomes in Patients Receiving Dual Antiplatelet Therapy after Percutaneous Coronary Intervention: A Propensity Score Analysis. Chin Med J (Engl) 2017; 130:2899-2905. [PMID: 29237921 PMCID: PMC5742916 DOI: 10.4103/0366-6999.220304] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Prior studies have reported controversial conclusions regarding the risk of adverse cardiovascular events in patients using proton-pump inhibitors (PPIs) combined with clopidogrel therapy, causing much uncertainty in clinical practice. We sought to evaluate the safety of PPIs use among high-risk cardiovascular patients who underwent percutaneous coronary intervention (PCI) in a long-term follow-up study. METHODS A total of 7868 consecutive patients who had undergone PCI and received dual antiplatelet therapy (DAPT) at a single center from January 2013 to December 2013 were enrolled. Adenosine diphosphate (ADP)-induced platelet aggregation inhibition was measured by modified thromboelastography (mTEG) in 5042 patients. Propensity score matching (PSM) was applied to control differing baseline factors. Cox proportional hazards regression was used to evaluate the 2-year major adverse cardiovascular and cerebrovascular events (MACCEs), as well as individual events, including all-cause death, myocardial infarction, unplanned target vessel revascularization, stent thrombosis, and stroke. RESULTS Among the whole cohort, 27.2% were prescribed PPIs. The ADP-induced platelet aggregation inhibition by mTEG was significantly lower in PPI users than that in non-PPI users (42.0 ± 30.9% vs. 46.4 ± 31.4%, t = 4.435, P < 0.001). Concomitant PPI use was not associated with increased MACCE through 2-year follow-up (12.7% vs. 12.5%, χ2 = 0.086, P = 0.769). Other endpoints showed no significant differences after multivariate adjustment, regardless of PSM. CONCLUSION In this large cohort of real-world patients, the combination of PPIs with DAPT was not associated with increased risk of MACCE in patients who underwent PCI at up to 2 years of follow-up.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Zhan Gao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Xiao-Fang Tang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Jing-Jing Xu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Yin Zhang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Li-Jian Gao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Jue Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Shu-Bin Qiao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Yue-Jin Yang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Run-Lin Gao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Bo Xu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Jin-Qing Yuan
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
929
|
Lüscher TF. Managing acute coronary syndromes: from novel biomarkers to mechanical support devices and DAPT in thrombocytopenia. Eur Heart J 2017. [DOI: 10.1093/eurheartj/ehx716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
930
|
De Luca L, Casella G, Rubboli A, Gonzini L, Lucci D, Boccanelli A, Chiarella F, Di Chiara A, De Servi S, Di Lenarda A, Di Pasquale G, Savonitto S. Recent trends in management and outcome of patients with acute coronary syndromes and atrial fibrillation. Int J Cardiol 2017; 248:369-375. [DOI: 10.1016/j.ijcard.2017.08.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/15/2017] [Accepted: 08/08/2017] [Indexed: 11/16/2022]
|
931
|
Ten Berg JM, Zwart B, van 't Hof AWJ, Liem A, Waltenberger J, de Winter RJ, Jukema JW. Optimal duration of dual antiplatelet therapy after percutaneous coronary intervention or after acute coronary syndrome : Practical lessons from a review. Neth Heart J 2017; 25:655-663. [PMID: 28762022 PMCID: PMC5691814 DOI: 10.1007/s12471-017-1023-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
To prevent recurrent ischaemic events, dual antiplatelet therapy (DAPT) is the standard of care after percutaneous coronary intervention and in the treatment of acute coronary syndrome. Recent evidence supports an adjusted DAPT duration in selected patients.The current paper aims to encourage cardiologists to actively search for patients benefiting from either shorter or prolonged duration DAPT and proposes an algorithm to identify patients who are likely to benefit from such an alternative strategy.Individualised DAPT duration should be considered in high-risk anatomic and/or clinical subgroups or in patients at increased haemorrhagic risk with low ischaemic risk. Both thrombotic and haemorrhagic risk should be assessed in all patients. In patients undergoing percutaneous coronary intervention, the interventional cardiologist could advise on the minimal duration of DAPT. However, in contrast to the minimum duration of DAPT for stent thrombosis prevention, longer duration DAPT is aimed at prevention of spontaneous myocardial infarction, and not at stent thrombosis, and thus the key to success is to treat the patient's overall thrombotic risk.The advice on the duration of DAPT must be documented in the patient's records and communicated with the treating physician and general practitioner. DAPT duration should be reassessed at least on a yearly basis.
Collapse
Affiliation(s)
- J M Ten Berg
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, The Netherlands.
| | - B Zwart
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - A W J van 't Hof
- Department of Cardiology, Isala Klinieken Zwolle, Zwolle, The Netherlands
| | - A Liem
- Department of Cardiology, Franciscus Gasthuis, Rotterdam, The Netherlands
| | - J Waltenberger
- Department of Cardiovascular Medicine, Universitätsklinikum Münster, Münster, Germany
| | - R J de Winter
- Department of Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - J W Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
932
|
Park SK, Jung DE, Jung SA, Kim WH, Bahk JH. Risk of non-cardiac surgery after percutaneous coronary intervention with drug-eluting stents. Sci Rep 2017; 7:16393. [PMID: 29180679 PMCID: PMC5704017 DOI: 10.1038/s41598-017-16672-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/16/2017] [Indexed: 02/02/2023] Open
Abstract
Elective non-cardiac surgery (NCS) should optimally be delayed one year after implantation of a drug-eluting stent (DES). Dual antiplatelet therapy or at least aspirin is recommended to be continued considering the relative risk of stent thrombosis especially during the 4 weeks after DES implantation. However, these recommendations were supported by insufficient evidence. We investigated predictors for postoperative major adverse cardiovascular and cerebral event (MACCE) in 1582 patients undergoing non-cardiac surgery after DES implantation. 96 patients (6.1%) developed postoperative MACCE. In the propensity score-matched analysis, aspirin maintenance was not associated with MACCE (odds ratio [OR] 0.78, 95% confidence interval [CI] 0.48-1.27, P = 0.320) and was associated with increased risk of major bleeding (OR 1.84, 95% CI 1.02-3.32, P = 0.044). When patients who underwent NCS within one month after DES implantation were matched with those who underwent NCS thereafter, the risk of MACCE was higher when surgery was done within 30 days after PCI (OR 2.21, 95% CI 1.05-4.66, P = 0.036). Maintenance of aspirin did not decrease MACCE after NCS in patients with DES and only increased the risk of major bleeding. NCS within one month after DES implantation was associated with higher incidence of MACCE. However, prospective trials are required to validate our results.
Collapse
Affiliation(s)
- Sun-Kyung Park
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dhong Eun Jung
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Ae Jung
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Won Ho Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Jae-Hyon Bahk
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
933
|
Lee JK, Wang IS, Huang CH, Chen YF, Huang NT, Lin CT. Pre-Clinical Tests of an Integrated CMOS Biomolecular Sensor for Cardiac Diseases Diagnosis. SENSORS 2017; 17:s17122733. [PMID: 29186872 PMCID: PMC5751442 DOI: 10.3390/s17122733] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 11/28/2022]
Abstract
Coronary artery disease and its related complications pose great threats to human health. In this work, we aim to clinically evaluate a CMOS field-effect biomolecular sensor for cardiac biomarkers, cardiac-specific troponin-I (cTnI), N-terminal prohormone brain natriuretic peptide (NT-proBNP), and interleukin-6 (IL-6). The CMOS biosensor is implemented via a standard commercialized 0.35 μm CMOS process. To validate the sensing characteristics, in buffer conditions, the developed CMOS biosensor has identified the detection limits of IL-6, cTnI, and NT-proBNP as being 45 pM, 32 pM, and 32 pM, respectively. In clinical serum conditions, furthermore, the developed CMOS biosensor performs a good correlation with an enzyme-linked immuno-sorbent assay (ELISA) obtained from a hospital central laboratory. Based on this work, the CMOS field-effect biosensor poses good potential for accomplishing the needs of a point-of-care testing (POCT) system for heart disease diagnosis.
Collapse
Affiliation(s)
- Jen-Kuang Lee
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan.
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10048, Taiwan.
- Telehealth Center, National Taiwan University Hospital, Taipei 10048, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 10048, Taiwan.
| | - I-Shun Wang
- Graduate Institute of Electronics Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Chi-Hsien Huang
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei 24301, Taiwan.
| | - Yih-Fan Chen
- Insisute of Biophotonics, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Nien-Tsu Huang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan.
| | - Chih-Ting Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Electronics Engineering, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
934
|
Ai S, He Z, Ding R, Wu F, Huang Z, Wang J, Huang S, Dai X, Zhang J, Chen J, Liu L, Wu Z, Liang C. Reduced Vitamin D Receptor on Circulating Endothelial Progenitor Cells: A New Risk Factor of Coronary Artery Diseases. J Atheroscler Thromb 2017; 25:410-421. [PMID: 29176261 PMCID: PMC5945554 DOI: 10.5551/jat.40808] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Aim: Endothelial progenitor cells (EPCs) are shown to participate in the pathological processes of atherosclerosis. While Vitamin D and its receptor axis might exert some effects on EPCs' function. But their exact relationship with clinical patients is still elusive, which inspired us to explore the potential association of vitamin D receptor (VDR) expression on circulating EPCs and serum vitamin D levels among patients with coronary artery disease (CAD). Methods: Two hundred patients with CAD after their admission to hospital and one hundred healthy controls were enrolled. Medical history data were retrieved and fresh blood samples were collected for flow cytometry analysis. VDR expressions on EPCs were evaluated according to the standardized protocol. Logistic regression analysis was used to investigate the potential risk factor of CAD. Results: CAD patients were found to have lower log10VDR-MFIs than those of control group, especially for patients with diabetes (p < 0.001). Log10VDR-MFIs were inversely correlated with glycated hemoglobin (R = −0.472, p < 0.001), and while EPCs challenged with high glucose had lower VDR expression. Multivariate logistic regression analysis revealed that lower log10VDR-MFIs were independently associated with the risk of CAD (OR = 0.055, p = 0.008). Conclusion: A significant decrease of VDR expression on circulating EPCs was observed among CAD patients, particularly among those also with diabetes. VDR expression on EPCs was independently negatively correlated with HbA1c and high glucose decreased EPCs' VDR expression. Low levels of VDR expression on circulating EPCs might serve as a potential risk factor of CAD.
Collapse
Affiliation(s)
- Sidi Ai
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Zhiqing He
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Ru Ding
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Feng Wu
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Zhigang Huang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Jiamei Wang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Shuaibo Huang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Xianliang Dai
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Jiayou Zhang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Jing Chen
- Department of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Linlin Liu
- Department of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Zonggui Wu
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| | - Chun Liang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University
| |
Collapse
|
935
|
Pinto DJP, Orwat MJ, Smith LM, Quan ML, Lam PYS, Rossi KA, Apedo A, Bozarth JM, Wu Y, Zheng JJ, Xin B, Toussaint N, Stetsko P, Gudmundsson O, Maxwell B, Crain EJ, Wong PC, Lou Z, Harper TW, Chacko SA, Myers JE, Sheriff S, Zhang H, Hou X, Mathur A, Seiffert DA, Wexler RR, Luettgen JM, Ewing WR. Discovery of a Parenteral Small Molecule Coagulation Factor XIa Inhibitor Clinical Candidate (BMS-962212). J Med Chem 2017; 60:9703-9723. [DOI: 10.1021/acs.jmedchem.7b01171] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Donald J. P. Pinto
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Michael J. Orwat
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Leon M. Smith
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Mimi L. Quan
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Patrick Y. S. Lam
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Karen A. Rossi
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Atsu Apedo
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Jeffrey M. Bozarth
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Yiming Wu
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joanna J. Zheng
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Baomin Xin
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Nathalie Toussaint
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Paul Stetsko
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Olafur Gudmundsson
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Brad Maxwell
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Earl J. Crain
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Pancras C. Wong
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Zhen Lou
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Timothy W. Harper
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Silvi A. Chacko
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joseph E. Myers
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Steven Sheriff
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Huiping Zhang
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Xiaoping Hou
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Arvind Mathur
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Dietmar A. Seiffert
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Ruth R. Wexler
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joseph M. Luettgen
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - William R. Ewing
- Research and Development, Bristol-Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543, United States
| |
Collapse
|
936
|
Prat M, Derumeaux H, Sailler L, Lapeyre-Mestre M, Moulis G. Positive predictive values of peripheral arterial and venous thrombosis codes in French hospital database. Fundam Clin Pharmacol 2017; 32:108-113. [DOI: 10.1111/fcp.12326] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 09/15/2017] [Accepted: 10/17/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Mandy Prat
- UMR 1027; INSERM; Faculté de Médecine; Université de Toulouse III; 37 allées Jules Guesde 31000 Toulouse France
- CIC 1436; Centre Hospitalier Universitaire de Toulouse; CHU Purpan, place du Dr Baylac, TSA 40031 31059 Toulouse France
| | - Hélène Derumeaux
- Département d'Information Médicale; Centre Hospitalier Universitaire de Toulouse; Hotel-Dieu, 2 rue Viguerie 31059 Toulouse France
| | - Laurent Sailler
- UMR 1027; INSERM; Faculté de Médecine; Université de Toulouse III; 37 allées Jules Guesde 31000 Toulouse France
- CIC 1436; Centre Hospitalier Universitaire de Toulouse; CHU Purpan, place du Dr Baylac, TSA 40031 31059 Toulouse France
- Service de Médecine Interne; Centre Hospitalier Universitaire de Toulouse; CHU Purpan, place du Dr Baylac, TSA 40031 31059 Toulouse France
| | - Maryse Lapeyre-Mestre
- UMR 1027; INSERM; Faculté de Médecine; Université de Toulouse III; 37 allées Jules Guesde 31000 Toulouse France
- CIC 1436; Centre Hospitalier Universitaire de Toulouse; CHU Purpan, place du Dr Baylac, TSA 40031 31059 Toulouse France
- Service de Pharmacologie Médicale et Clinique; Faculté de Médecine; Centre Hospitalier Universitaire de Toulouse; 37 allées Jules Guesde 31000 Toulouse France
| | - Guillaume Moulis
- UMR 1027; INSERM; Faculté de Médecine; Université de Toulouse III; 37 allées Jules Guesde 31000 Toulouse France
- CIC 1436; Centre Hospitalier Universitaire de Toulouse; CHU Purpan, place du Dr Baylac, TSA 40031 31059 Toulouse France
- Service de Médecine Interne; Centre Hospitalier Universitaire de Toulouse; CHU Purpan, place du Dr Baylac, TSA 40031 31059 Toulouse France
| |
Collapse
|
937
|
Angiolillo DJ, Goodman SG, Bhatt DL, Eikelboom JW, Price MJ, Moliterno DJ, Cannon CP, Tanguay JF, Granger CB, Mauri L, Holmes DR, Gibson CM, Faxon DP. Antithrombotic Therapy in Patients With Atrial Fibrillation Undergoing Percutaneous Coronary Intervention: A North American Perspective-2016 Update. Circ Cardiovasc Interv 2017; 9:CIRCINTERVENTIONS.116.004395. [PMID: 27803042 DOI: 10.1161/circinterventions.116.004395] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The optimal antithrombotic treatment regimen for patients with atrial fibrillation undergoing percutaneous coronary intervention with stent implantation is an emerging clinical problem. Currently, there is limited evidenced-based data on the optimal antithrombotic treatment regimen, including antiplatelet and anticoagulant therapies, for these high-risk patients with practice guidelines, thus, providing limited recommendations. Over the past years, expert consensus documents have provided guidance to clinicians on how to manage patients with atrial fibrillation undergoing percutaneous coronary intervention. Given the recent advancements in the field, the current document provides an updated opinion of selected North American experts from the United States and Canada on the treatment of patients with atrial fibrillation undergoing percutaneous coronary intervention. In particular, this document provides the current views on (1) embolic/stroke risk, (2) ischemic/thrombotic cardiac risk, and (3) bleeding risk, which are pivotal for discerning the choice of antithrombotic therapy. In addition, we describe the recent advances in pharmacology, stent designs, and clinical trials relevant to the field. Ultimately, we provide expert consensus-derived recommendations, using a pragmatic approach, on the management of patients with atrial fibrillation undergoing percutaneous coronary intervention.
Collapse
Affiliation(s)
- Dominick J Angiolillo
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.).
| | - Shaun G Goodman
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - Deepak L Bhatt
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - John W Eikelboom
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - Matthew J Price
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - David J Moliterno
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - Christopher P Cannon
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - Jean-Francois Tanguay
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - Christopher B Granger
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - Laura Mauri
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - David R Holmes
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - C Michael Gibson
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| | - David P Faxon
- From the Division of Cardiology, University of Florida College of Medicine-Jacksonville (D.J.A.); St Michael's Hospital, University of Toronto, and the Canadian Heart Research Centre; Canadian VIGOUR Centre, University of Alberta, Edmonton (S.G.G.); Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, MA (D.L.B., D.P.F.); Department of Medicine, Population Health Research Institute, Thrombosis & Atherosclerosis Research Institute, Hamilton, ON, Canada (J.W.E.); Division of Cardiovascular Diseases, Scripps Clinic, La Jolla CA (M.J.P.); Division of Cardiovascular Medicine and Gill Heart Institute, University of Kentucky, Lexington (D.J.M.); Brigham and Women's Hospital, Harvard Clinical Research Institute, Harvard Medical School, Boston, MA (C.P.C., L.M.); Department of Medicine, Montreal Heart Institute, Université de Montréal, QC, Canada (J.-F.T.); Duke Clinical Research Institute, Duke University, Durham, NC (C.B.G.); Mayo Clinic, Rochester, MN (D.R.H.); and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (C.M.G.)
| |
Collapse
|
938
|
Goli RR, Contractor MM, Nathan A, Tuteja S, Kobayashi T, Giri J. Antiplatelet Therapy for Secondary Prevention of Vascular Disease Complications. Curr Atheroscler Rep 2017; 19:56. [DOI: 10.1007/s11883-017-0698-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
939
|
Calabrò P, Gragnano F, De Luca L. Dual Antiplatelet Therapy Duration in Conservatively Managed Patients with Acute Coronary Syndrome: Any News? Am J Med 2017; 130:e515. [PMID: 29073976 DOI: 10.1016/j.amjmed.2017.06.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 06/26/2017] [Indexed: 11/18/2022]
Affiliation(s)
- Paolo Calabrò
- Division of Cardiology, Department of Cardiothoracic and Respiratory Sciences, University of Campania "Luigi Vanvitelli", A.O. dei Colli Monaldi Hospital, L. Bianchi 1,Naples 80131, Italy
| | - Felice Gragnano
- Division of Cardiology, Department of Cardiothoracic and Respiratory Sciences, University of Campania "Luigi Vanvitelli", A.O. dei Colli Monaldi Hospital, L. Bianchi 1,Naples 80131, Italy
| | - Leonardo De Luca
- Division of Cardiology, Laboratory of Interventional Cardiology, San Giovanni Evangelista Hospital, Via Parrozzani 3,Tivoli, Rome 00019, Italy
| |
Collapse
|
940
|
Cavallari LH, Lee CR, Beitelshees AL, Cooper-DeHoff RM, Duarte JD, Voora D, Kimmel SE, McDonough CW, Gong Y, Dave CV, Pratt VM, Alestock TD, Anderson RD, Alsip J, Ardati AK, Brott BC, Brown L, Chumnumwat S, Clare-Salzler MJ, Coons JC, Denny JC, Dillon C, Elsey AR, Hamadeh IS, Harada S, Hillegass WB, Hines L, Horenstein RB, Howell LA, Jeng LJB, Kelemen MD, Lee YM, Magvanjav O, Montasser M, Nelson DR, Nutescu EA, Nwaba DC, Pakyz RE, Palmer K, Peterson JF, Pollin TI, Quinn AH, Robinson SW, Schub J, Skaar TC, Smith DM, Sriramoju VB, Starostik P, Stys TP, Stevenson JM, Varunok N, Vesely MR, Wake DT, Weck KE, Weitzel KW, Wilke RA, Willig J, Zhao RY, Kreutz RP, Stouffer GA, Empey PE, Limdi NA, Shuldiner AR, Winterstein AG, Johnson JA. Multisite Investigation of Outcomes With Implementation of CYP2C19 Genotype-Guided Antiplatelet Therapy After Percutaneous Coronary Intervention. JACC Cardiovasc Interv 2017; 11:181-191. [PMID: 29102571 DOI: 10.1016/j.jcin.2017.07.022] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/07/2017] [Accepted: 07/11/2017] [Indexed: 01/14/2023]
Abstract
OBJECTIVES This multicenter pragmatic investigation assessed outcomes following clinical implementation of CYP2C19 genotype-guided antiplatelet therapy after percutaneous coronary intervention (PCI). BACKGROUND CYP2C19 loss-of-function alleles impair clopidogrel effectiveness after PCI. METHODS After clinical genotyping, each institution recommended alternative antiplatelet therapy (prasugrel, ticagrelor) in PCI patients with a loss-of-function allele. Major adverse cardiovascular events (defined as myocardial infarction, stroke, or death) within 12 months of PCI were compared between patients with a loss-of-function allele prescribed clopidogrel versus alternative therapy. Risk was also compared between patients without a loss-of-function allele and loss-of-function allele carriers prescribed alternative therapy. Cox regression was performed, adjusting for group differences with inverse probability of treatment weights. RESULTS Among 1,815 patients, 572 (31.5%) had a loss-of-function allele. The risk for major adverse cardiovascular events was significantly higher in patients with a loss-of-function allele prescribed clopidogrel versus alternative therapy (23.4 vs. 8.7 per 100 patient-years; adjusted hazard ratio: 2.26; 95% confidence interval: 1.18 to 4.32; p = 0.013). Similar results were observed among 1,210 patients with acute coronary syndromes at the time of PCI (adjusted hazard ratio: 2.87; 95% confidence interval: 1.35 to 6.09; p = 0.013). There was no difference in major adverse cardiovascular events between patients without a loss-of-function allele and loss-of-function allele carriers prescribed alternative therapy (adjusted hazard ratio: 1.14; 95% confidence interval: 0.69 to 1.88; p = 0.60). CONCLUSIONS These data from real-world observations demonstrate a higher risk for cardiovascular events in patients with a CYP2C19 loss-of-function allele if clopidogrel versus alternative therapy is prescribed. A future randomized study of genotype-guided antiplatelet therapy may be of value.
Collapse
Affiliation(s)
- Larisa H Cavallari
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida.
| | - Craig R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida; Department of Medicine, Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida
| | - Julio D Duarte
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois
| | - Deepak Voora
- Department of Medicine, Center for Applied Genomics & Precision Medicine, Duke University, Durham, North Carolina
| | - Stephen E Kimmel
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Caitrin W McDonough
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - Chintan V Dave
- Department of Pharmaceutical Outcomes and Policy, University of Florida, Gainesville, Florida
| | - Victoria M Pratt
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - R David Anderson
- Department of Medicine, Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida
| | - Jorge Alsip
- Division of Cardiovascular Sciences, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amer K Ardati
- Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Brigitta C Brott
- Division of Cardiovascular Sciences, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lawrence Brown
- Veterans Administration Medical Center, Baltimore, Maryland
| | - Supatat Chumnumwat
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois
| | - Michael J Clare-Salzler
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - James C Coons
- Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Joshua C Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Chrisly Dillon
- Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amanda R Elsey
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - Issam S Hamadeh
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - Shuko Harada
- Department of Pathology and Hugh Kaul Personalized Medicine Institute, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - William B Hillegass
- Heart South Cardiovascular Group, Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lindsay Hines
- Department of Neuropsychology, University of North Dakota, Fargo, North Dakota
| | | | - Lucius A Howell
- Division of Cardiology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Linda J B Jeng
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Mark D Kelemen
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Yee Ming Lee
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois
| | - Oyunbileg Magvanjav
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - May Montasser
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - David R Nelson
- College of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, Florida
| | - Edith A Nutescu
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois; Department of Pharmacy Systems, Outcomes and Policy and Center for Pharmacoepidemiology and Pharmacoeconomic Research, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois
| | - Devon C Nwaba
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Ruth E Pakyz
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Kathleen Palmer
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Josh F Peterson
- Departments of Biomedical Informatics and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Toni I Pollin
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Alison H Quinn
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois
| | - Shawn W Robinson
- Department of Medicine, University of Maryland, Baltimore, Maryland; Veterans Administration Medical Center, Baltimore, Maryland
| | - Jamie Schub
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Todd C Skaar
- Department of Medicine, Krannert Institute of Cardiology & Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana
| | - D Max Smith
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - Vindhya B Sriramoju
- Division of Cardiology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Petr Starostik
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Tomasz P Stys
- Department of Medicine, University of South Dakota, Sanford School of Medicine, Sioux Falls, South Dakota
| | - James M Stevenson
- Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Nicholas Varunok
- Division of Cardiology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mark R Vesely
- Department of Medicine, University of Maryland, Baltimore, Maryland; Veterans Administration Medical Center, Baltimore, Maryland
| | - Dyson T Wake
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - Karen E Weck
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kristin W Weitzel
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - Russell A Wilke
- Department of Medicine, University of South Dakota, Sanford School of Medicine, Sioux Falls, South Dakota
| | - James Willig
- Division of Cardiovascular Sciences, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Richard Y Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rolf P Kreutz
- Department of Medicine, Krannert Institute of Cardiology & Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana
| | - George A Stouffer
- Division of Cardiology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Philip E Empey
- Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Nita A Limdi
- Department of Neurology and Hugh Kaul Personalized Medicine Institute, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Alan R Shuldiner
- Department of Medicine, University of Maryland, Baltimore, Maryland
| | - Almut G Winterstein
- Department of Pharmaceutical Outcomes and Policy, University of Florida, Gainesville, Florida; Department of Epidemiology, Colleges of Medicine and Public Health and Health Professions, University of Florida, Gainesville, Florida
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida; Department of Medicine, Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida
| | | |
Collapse
|
941
|
Agarwal N, Jain A, Mahmoud AN, Bishnoi R, Golwala H, Karimi A, Mojadidi MK, Garg J, Gupta T, Patel NK, Wayangankar S, Anderson RD. Safety and Efficacy of Dual Versus Triple Antithrombotic Therapy in Patients Undergoing Percutaneous Coronary Intervention. Am J Med 2017; 130:1280-1289. [PMID: 28460853 DOI: 10.1016/j.amjmed.2017.03.057] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 03/28/2017] [Accepted: 03/28/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Choosing an antithrombotic regimen after coronary intervention in patients with concomitant indication for anticoagulation is a challenge commonly encountered by clinicians. METHODS We performed a meta-analysis of observational studies and randomized, controlled trials comparing outcomes of triple therapy (dual antiplatelet therapy and anticoagulant) with dual therapy (single antiplatelet therapy and anticoagulant) in patients taking long-term anticoagulants after percutaneous coronary intervention. Major bleeding was the primary outcome. Random effects overall risk ratios (RRs) were calculated using the DerSimonian and Laird model. RESULTS Nine observational studies and 2 randomized controlled trials with a total of 7276 patients met our selection criteria. At a mean follow-up of 10.8 months major bleeding was higher in the triple therapy cohort compared with dual therapy (6.6% vs 3.8%; RR 1.54; 95% confidence interval [CI], 1.2-1.98; P <.01). No difference was observed between the 2 groups for all-cause mortality (RR 0.98; 95% CI, 0.68-1.43; P = .93), major adverse cardiac events (RR 1.03; 95% CI, 0.8-1.32; P = .83), thromboembolic events (RR 1.02; 95% CI, 0.49-2.10; P = .96), myocardial infarction (RR 0.85; 95% CI, 0.67-1.09; P = .21), stent thrombosis (RR 0.77; 95% CI, 0.46-1.3; P = .33), and target vessel revascularization (RR 0.87; 95% CI, 0.66-1.15; P = .33). CONCLUSION In patients receiving anticoagulant therapy, a strategy of single antiplatelet therapy confers a benefit of less major bleeding with no difference in all-cause mortality, cardiovascular mortality, major adverse cardiac events, myocardial infarction, stent thrombosis, or thromboembolic event rate compared with dual antiplatelet therapy.
Collapse
Affiliation(s)
- Nayan Agarwal
- Department of Medicine, University of Florida, Gainesville
| | - Ankur Jain
- Department of Medicine, University of Florida, Gainesville
| | | | - Rohit Bishnoi
- Department of Medicine, University of Florida, Gainesville
| | - Harsh Golwala
- Department of Medicine, Brigham and Women's Hospital, Boston, Mass
| | - Ashkan Karimi
- Department of Medicine, University of Florida, Gainesville
| | | | - Jalaj Garg
- Department of Medicine, Lehigh Valley Hospital, Allentown, Pa
| | - Tanush Gupta
- Department of Medicine, Montefiore Medical Centre, Albert Einstein College of Medicine, Bronx, NY
| | - Nimesh Kirit Patel
- Department of Medicine, Virginia Commonwealth University Health System, Richmond
| | | | | |
Collapse
|
942
|
Meyer A, Gross N, Teng M. AHNS Series: Do you know your guidelines? Perioperative antithrombotic management in head and neck surgery. Head Neck 2017; 40:182-191. [PMID: 29044795 DOI: 10.1002/hed.24927] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 07/20/2017] [Indexed: 12/16/2022] Open
Abstract
Head and neck surgeons are commonly faced with surgical patients who have underlying medical problems requiring antithrombotic therapy. It is difficult to achieve a balance between minimizing the risk of thromboembolism and hemorrhage in the perioperative period. Data from randomized, controlled trials are limited, and procedure-specific bleed rates are also difficult to pinpoint. The decision is made more difficult when patients with moderate-to-high risk for thromboembolic events undergo procedures that are high risk for bleeding. This is true for many head and neck oncologic surgeries. Furthermore, although elective procedures may be delayed for optimization of antithrombotic medication, emergent procedures cannot. Head and neck surgery often represents the most challenging of all these circumstances, given the potential risk of airway compromise from bleeding after head and neck surgery.
Collapse
Affiliation(s)
- Annika Meyer
- Department of Otolaryngology - Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Neil Gross
- MD Anderson Cancer Center, Head and Neck Surgery, Houston, Texas
| | - Marita Teng
- Department of Otolaryngology - Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
943
|
Cannon CP, Bhatt DL, Oldgren J, Lip GYH, Ellis SG, Kimura T, Maeng M, Merkely B, Zeymer U, Gropper S, Nordaby M, Kleine E, Harper R, Manassie J, Januzzi JL, Ten Berg JM, Steg PG, Hohnloser SH. Dual Antithrombotic Therapy with Dabigatran after PCI in Atrial Fibrillation. N Engl J Med 2017; 377:1513-1524. [PMID: 28844193 DOI: 10.1056/nejmoa1708454] [Citation(s) in RCA: 967] [Impact Index Per Article: 120.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Triple antithrombotic therapy with warfarin plus two antiplatelet agents is the standard of care after percutaneous coronary intervention (PCI) for patients with atrial fibrillation, but this therapy is associated with a high risk of bleeding. METHODS In this multicenter trial, we randomly assigned 2725 patients with atrial fibrillation who had undergone PCI to triple therapy with warfarin plus a P2Y12 inhibitor (clopidogrel or ticagrelor) and aspirin (for 1 to 3 months) (triple-therapy group) or dual therapy with dabigatran (110 mg or 150 mg twice daily) plus a P2Y12 inhibitor (clopidogrel or ticagrelor) and no aspirin (110-mg and 150-mg dual-therapy groups). Outside the United States, elderly patients (≥80 years of age; ≥70 years of age in Japan) were randomly assigned to the 110-mg dual-therapy group or the triple-therapy group. The primary end point was a major or clinically relevant nonmajor bleeding event during follow-up (mean follow-up, 14 months). The trial also tested for the noninferiority of dual therapy with dabigatran (both doses combined) to triple therapy with warfarin with respect to the incidence of a composite efficacy end point of thromboembolic events (myocardial infarction, stroke, or systemic embolism), death, or unplanned revascularization. RESULTS The incidence of the primary end point was 15.4% in the 110-mg dual-therapy group as compared with 26.9% in the triple-therapy group (hazard ratio, 0.52; 95% confidence interval [CI], 0.42 to 0.63; P<0.001 for noninferiority; P<0.001 for superiority) and 20.2% in the 150-mg dual-therapy group as compared with 25.7% in the corresponding triple-therapy group, which did not include elderly patients outside the United States (hazard ratio, 0.72; 95% CI, 0.58 to 0.88; P<0.001 for noninferiority). The incidence of the composite efficacy end point was 13.7% in the two dual-therapy groups combined as compared with 13.4% in the triple-therapy group (hazard ratio, 1.04; 95% CI, 0.84 to 1.29; P=0.005 for noninferiority). The rate of serious adverse events did not differ significantly among the groups. CONCLUSIONS Among patients with atrial fibrillation who had undergone PCI, the risk of bleeding was lower among those who received dual therapy with dabigatran and a P2Y12 inhibitor than among those who received triple therapy with warfarin, a P2Y12 inhibitor, and aspirin. Dual therapy was noninferior to triple therapy with respect to the risk of thromboembolic events. (Funded by Boehringer Ingelheim; RE-DUAL PCI ClinicalTrials.gov number, NCT02164864 .).
Collapse
Affiliation(s)
- Christopher P Cannon
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Deepak L Bhatt
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Jonas Oldgren
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Gregory Y H Lip
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Stephen G Ellis
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Takeshi Kimura
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Michael Maeng
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Bela Merkely
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Uwe Zeymer
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Savion Gropper
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Matias Nordaby
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Eva Kleine
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Ruth Harper
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Jenny Manassie
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - James L Januzzi
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Jurrien M Ten Berg
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - P Gabriel Steg
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| | - Stefan H Hohnloser
- From the Baim Institute for Clinical Research (C.P.C., J.L.J.), Brigham and Women's Hospital, Heart and Vascular Center, and Harvard Medical School (C.P.C., D.L.B.), and the Cardiology Division, Massachusetts General Hospital, and Harvard Medical School (J.L.J.) - all in Boston; Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden (J.O.); the Institute of Cardiovascular Sciences, University of Birmingham, Birmingham (G.Y.H.L.), Boehringer Ingelheim, Bracknell (R.H., J.M.), and Imperial College, London, London (P.G.S.) - all in the United Kingdom; Cleveland Clinic, Cleveland (S.G.E.); Kyoto University, Department of Cardiovascular Medicine, Kyoto, Japan (T.K.); Aarhus University Hospital, Skejby, Denmark (M.M.); University Heart and Vascular Center, Budapest, Hungary (B.M.); Klinikum der Stadt Ludwigshafen am Rhein, Medizinische Klinik B, Ludwigshafen (U.Z.), Boehringer Ingelheim, Ingelheim (S.G., M.N., E.K.), and Johann Wolfgang Goethe University, Department of Medicine, Division of Cardiology, Frankfurt am Main (S.H.H.) - all in Germany; St. Antonius Ziekenhuis, Nieuwegein, the Netherlands (J.M.B.); and the French Alliance for Cardiovascular Trials, F-CRIN Network, DHU FIRE, Université Paris Diderot, INSERM Unité 1148, and Hôpital Bichat Assistance Publique, Paris (P.G.S.)
| |
Collapse
|
944
|
Patterns and levels of platelet glycosylation in patients with coronary heart disease and type 2 diabetes mellitus. J Thromb Thrombolysis 2017; 45:56-65. [DOI: 10.1007/s11239-017-1573-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
945
|
9-Month Clinical and Angiographic Outcomes of the COBRA Polyzene-F NanoCoated Coronary Stent System. JACC Cardiovasc Interv 2017; 10:160-167. [PMID: 28104210 DOI: 10.1016/j.jcin.2016.10.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/27/2016] [Accepted: 10/28/2016] [Indexed: 11/23/2022]
Abstract
OBJECTIVES The aim of this study was to assess the safety and effectiveness of the COBRA Polyzene-F NanoCoated Coronary Stent System (CeloNova Biosciences, San Antonio, Texas) for the treatment of de novo coronary artery lesions. BACKGROUND Polyzene-F-coated coronary stents have shown reduced thrombogenicity and inflammation in preclinical studies. METHODS Patients with de novo coronary artery lesions meeting eligibility criteria were enrolled in a nonrandomized, prospective clinical trial. The primary endpoint was target vessel failure (TVF) (defined as a composite of cardiac death, myocardial infarction, or clinically driven target vessel revascularization) at 9 months. A pre-specified subset was planned for routine repeat angiographic follow-up at 9 months. The powered secondary endpoint was mean late lumen loss (LL). The comparator was a performance goal derived from meta-analysis of historical bare-metal stent trials of 19.62% for TVF and 1.1 mm for LL. Other secondary endpoints were clinically driven target lesion revascularization and definite or probable stent thrombosis. RESULTS Of 296 enrolled patients, 287 (97%) completed primary endpoint analysis; 130 were planned for angiographic follow-up and 115 (88%) completed. At 9 months, TVF had occurred in 33 patients (11.5%; upper 95% confidence boundary: 15.07%), including 1 (0.3%) cardiac death, 20 (7.0%) myocardial infarctions (17 periprocedural), and 17 (5.9%) target vessel revascularizations. LL was 0.84 ± 0.48 mm (upper 95% confidence boundary: 0.92). Target lesion revascularization occurred in 13 patients (4.6%). There were no stent thrombosis events. CONCLUSIONS The COBRA Polyzene-F stent met performance goals for TVF and LL at 9 months. There was an excellent safety profile, with infrequent late myocardial infarction and no stent thrombosis.
Collapse
|
946
|
Arbel Y, Bennell MC, Goodman SG, Wijeysundera HC. Cost-Effectiveness of Different Durations of Dual-Antiplatelet Use After Percutaneous Coronary Intervention. Can J Cardiol 2017; 34:31-37. [PMID: 29275879 DOI: 10.1016/j.cjca.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 10/01/2017] [Accepted: 10/02/2017] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND There is uncertainty regarding the optimal duration of dual-antiplatelet therapy (DAPT) after percutaneous coronary intervention (PCI). Our goal was to evaluate the cost-effectiveness of different durations of DAPT. METHODS We created a probabilistic patient-level Markov microsimulation model to assess the discounted lifetime costs and quality-adjusted life years (QALYs) of short duration (3-6 months: short-duration group) vs standard therapy (12 months: standard-duration group) vs prolonged therapy (30-36 months: long-duration group) in patients undergoing PCI. RESULTS The majority of patients in the model underwent PCI for stable angina (47.1%) with second-generation drug-eluting stents (62%) and were receiving clopidogrel (83.6%). Short-duration DAPT was the most effective strategy (7.163 ± 1.098 QALYs) compared with standard-duration DAPT (7.161 ± 1.097 QALYs) and long-duration DAPT (7.156 ± 1.097 QALYs). However, the magnitude of these differences was very small. Similarly, the average discounted lifetime cost was CAN$24,859 ± $6533 for short duration, $25,045 ± $6533 for standard duration, and $25,046 ± $6548 for long duration. Thus, in the base-case analysis, short duration was dominant, being more effective and less expensive. However, there was a moderate degree of uncertainty, because short duration was the preferred option in only ∼ 55% of simulations at a willingness to pay threshold of $50,000. CONCLUSIONS Based on a stable angina cohort receiving clopidogrel with second-generation stents, a short duration of DAPT was marginally better. However, the differences are minimal, and decisions about duration of therapy should be driven by clinical data, patient risk of adverse events, including bleeding, and cardiovascular events.
Collapse
Affiliation(s)
- Yaron Arbel
- Department of Cardiology, Tel Aviv Medical Center, Tel Aviv, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Schulich Heart Centre, Division of Cardiology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Maria C Bennell
- Schulich Heart Centre, Division of Cardiology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Shaun G Goodman
- St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Harindra C Wijeysundera
- Schulich Heart Centre, Division of Cardiology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences (ICES), Toronto, Ontario, Canada; Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
947
|
Russo JJ, Goodman SG, Bagai A, Déry JP, Tan MK, Fisher HN, Zhang X, Zhu YE, Welsh RC, Siega AD, Kokis A, Wong BYL, Henderson M, Lutchmedial S, Lavi S, Mehta SR, Yan AT. Duration of dual antiplatelet therapy and associated outcomes following percutaneous coronary intervention for acute myocardial infarction: contemporary practice insights from the Canadian Observational Antiplatelet Study. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2017; 3:303-311. [PMID: 29044393 DOI: 10.1093/ehjqcco/qcw051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Indexed: 12/22/2022]
Abstract
Aims There is a paucity of real-world, contemporary data of practice patterns and clinical outcomes following dual-antiplatelet therapy (DAPT) in acute myocardial infarction (AMI) patients treated with percutaneous coronary intervention (PCI). Methods and results The Canadian Observational Antiplatelet Study was a prospective, multicentre, cohort study examining adenosine diphosphate receptor antagonist use following PCI for AMI. We compared practice patterns, patient characteristics, and clinical outcomes in relation to DAPT duration (<6 weeks, 6 weeks to <6 months, 6 to <12, and ≥12 months). The primary outcome was the composite of non-fatal AMI, unplanned coronary revascularization, stent thrombosis, new or worsening heart failure, cardiogenic shock, or stroke. We identified 2034 patients with AMI treated with PCI. DAPT duration was <6 weeks in 5.2% of patients; 6 weeks to <6 months in 7.0%; 6 to <12 months in 12.6%; and ≥12 months in 75.3%. Patients who discontinued DAPT early had higher GRACE risk scores. Overall, mortality rate at 15 months was 2.5%. Compared with a duration of DAPT of ≥12 months, discontinuation of DAPT <6 weeks (P < 0.0001) and 6 weeks to <6 months (P = 0.02), but not 6 months to <12 months (P = 0.06), were independently associated with a higher incidence of the primary outcome among survivors. Conclusion One-in-four patients with AMI treated with PCI discontinued DAPT prior to the guideline-recommended 12-month duration. Patients in whom DAPT was discontinued early were at higher baseline risk and had higher rates of non-fatal ischaemic events during follow up.
Collapse
Affiliation(s)
- Juan J Russo
- Division of Cardiology, Terrence Donnelly Heart Centre, St Michael's Hospital, University of Toronto, 30 Bond Street, Donnelly 6-030, Toronto, ON M5B 1W8, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Shaun G Goodman
- Division of Cardiology, Terrence Donnelly Heart Centre, St Michael's Hospital, University of Toronto, 30 Bond Street, Donnelly 6-030, Toronto, ON M5B 1W8, Canada
- Canadian Heart Research Centre, Toronto, ON, Canada
| | - Akshay Bagai
- Division of Cardiology, Terrence Donnelly Heart Centre, St Michael's Hospital, University of Toronto, 30 Bond Street, Donnelly 6-030, Toronto, ON M5B 1W8, Canada
| | - Jean-Pierre Déry
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec City, QC, Canada
| | - Mary K Tan
- Canadian Heart Research Centre, Toronto, ON, Canada
| | | | | | | | - Robert C Welsh
- The Mazankowski Alberta Heart Institute, University of Alberta, VIGOUR Centre, Edmonton, AB, Canada
| | | | - Andre Kokis
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | | | - Mark Henderson
- Thunder Bay Regional Health Sciences Centre, Thunder Bay, ON, Canada
| | - Sohrab Lutchmedial
- New Brunswick Heart Centre, CardioVascular Research New Brunswick, Saint John, NB, Canada
| | - Shahar Lavi
- London Health Sciences Centre, Western University, London, ON, Canada
| | - Shamir R Mehta
- Hamilton Health Sciences, McMaster University, Population Health Research Institute, Hamilton, ON, Canada
| | - Andrew T Yan
- Division of Cardiology, Terrence Donnelly Heart Centre, St Michael's Hospital, University of Toronto, 30 Bond Street, Donnelly 6-030, Toronto, ON M5B 1W8, Canada
| |
Collapse
|
948
|
Michel J. Turning Guidelines Into Outcomes. Proc (Bayl Univ Med Cent) 2017; 30:413-414. [DOI: 10.1080/08998280.2017.11930209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Jeffrey Michel
- Scott and White Heart & Vascular Institute Temple, Texas
| |
Collapse
|
949
|
Jneid H, Addison D, Bhatt DL, Fonarow GC, Gokak S, Grady KL, Green LA, Heidenreich PA, Ho PM, Jurgens CY, King ML, Kumbhani DJ, Pancholy S. 2017 AHA/ACC Clinical Performance and Quality Measures for Adults With ST-Elevation and Non–ST-Elevation Myocardial Infarction: A Report of the American College of Cardiology/American Heart Association Task Force on Performance Measures. Circ Cardiovasc Qual Outcomes 2017; 10:HCQ.0000000000000032. [DOI: 10.1161/hcq.0000000000000032] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
950
|
Regan DW, Kashiwagi D, Dougan B, Sundsted K, Mauck K. Update in perioperative medicine: practice changing evidence published in 2016. Hosp Pract (1995) 2017; 45:158-164. [PMID: 28749248 DOI: 10.1080/21548331.2017.1359060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/20/2017] [Indexed: 06/07/2023]
Abstract
This summary reviews 18 key articles published in 2016 which have significant practice implications for the perioperative medical care of surgical patients. Due to the multi-disciplinary nature of the practice of perioperative medicine, important new evidence is published in journals representing a variety of medical and surgical specialties. Keeping current with the evidence that drives best practice in perioperative medicine is therefore challenging. We set out to identify, critically review, and summarize key evidence which has the most potential for practice change. We integrated the new evidence into the existing body of medical knowledge and identified practical implications for real world patient care. The articles address issues related to anticoagulation, transfusion threshold, immunosuppressive medications, postoperative delirium, myocardial injury after noncardiac surgery, postoperative pain management, perioperative management of antihypertensives, perioperative fasting, and perioperative diabetic control.
Collapse
Affiliation(s)
- Dennis W Regan
- a General Internal Medicine , Mayo Clinic , Rochester , MN , USA
| | | | - Brian Dougan
- a General Internal Medicine , Mayo Clinic , Rochester , MN , USA
| | - Karna Sundsted
- a General Internal Medicine , Mayo Clinic , Rochester , MN , USA
| | - Karen Mauck
- a General Internal Medicine , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|