Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Witold Rużyłło is active.

Publication


Featured researches published by Witold Rużyłło.


The New England Journal of Medicine | 2013

Edoxaban versus Warfarin in Patients with Atrial Fibrillation

Robert P. Giugliano; Christian T. Ruff; Eugene Braunwald; Sabina A. Murphy; Stephen D. Wiviott; Jonathan L. Halperin; Albert L. Waldo; Michael D. Ezekowitz; Jeffrey I. Weitz; Witold Rużyłło; Mikhail Ruda; Yukihiro Koretsune; Joshua Betcher; Minggao Shi; Laura T. Grip; Shirali P. Patel; Indravadan Patel; James J. Hanyok; Michele Mercuri; Elliott M. Antman; Abstr Act

BACKGROUND Edoxaban is a direct oral factor Xa inhibitor with proven antithrombotic effects. The long-term efficacy and safety of edoxaban as compared with warfarin in patients with atrial fibrillation is not known. METHODS We conducted a randomized, double-blind, double-dummy trial comparing two once-daily regimens of edoxaban with warfarin in 21,105 patients with moderate-to-high-risk atrial fibrillation (median follow-up, 2.8 years). The primary efficacy end point was stroke or systemic embolism. Each edoxaban regimen was tested for noninferiority to warfarin during the treatment period. The principal safety end point was major bleeding. RESULTS The annualized rate of the primary end point during treatment was 1.50% with warfarin (median time in the therapeutic range, 68.4%), as compared with 1.18% with high-dose edoxaban (hazard ratio, 0.79; 97.5% confidence interval [CI], 0.63 to 0.99; P<0.001 for noninferiority) and 1.61% with low-dose edoxaban (hazard ratio, 1.07; 97.5% CI, 0.87 to 1.31; P=0.005 for noninferiority). In the intention-to-treat analysis, there was a trend favoring high-dose edoxaban versus warfarin (hazard ratio, 0.87; 97.5% CI, 0.73 to 1.04; P=0.08) and an unfavorable trend with low-dose edoxaban versus warfarin (hazard ratio, 1.13; 97.5% CI, 0.96 to 1.34; P=0.10). The annualized rate of major bleeding was 3.43% with warfarin versus 2.75% with high-dose edoxaban (hazard ratio, 0.80; 95% CI, 0.71 to 0.91; P<0.001) and 1.61% with low-dose edoxaban (hazard ratio, 0.47; 95% CI, 0.41 to 0.55; P<0.001). The corresponding annualized rates of death from cardiovascular causes were 3.17% versus 2.74% (hazard ratio, 0.86; 95% CI, 0.77 to 0.97; P=0.01), and 2.71% (hazard ratio, 0.85; 95% CI, 0.76 to 0.96; P=0.008), and the corresponding rates of the key secondary end point (a composite of stroke, systemic embolism, or death from cardiovascular causes) were 4.43% versus 3.85% (hazard ratio, 0.87; 95% CI, 0.78 to 0.96; P=0.005), and 4.23% (hazard ratio, 0.95; 95% CI, 0.86 to 1.05; P=0.32). CONCLUSIONS Both once-daily regimens of edoxaban were noninferior to warfarin with respect to the prevention of stroke or systemic embolism and were associated with significantly lower rates of bleeding and death from cardiovascular causes. (Funded by Daiichi Sankyo Pharma Development; ENGAGE AF-TIMI 48 ClinicalTrials.gov number, NCT00781391.).


The New England Journal of Medicine | 2015

Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes

Christopher P. Cannon; Michael A. Blazing; Robert P. Giugliano; Amy McCagg; Jennifer A. White; Harald Darius; Basil S. Lewis; Ton Oude Ophuis; J. Wouter Jukema; Gaetano M. De Ferrari; Witold Rużyłło; Paul De Lucca; Erin A. Bohula; Craig J. Reist; Stephen D. Wiviott; Andrew M. Tershakovec; Thomas Musliner; Eugene Braunwald; Robert M. Califf

BACKGROUND Statin therapy reduces low-density lipoprotein (LDL) cholesterol levels and the risk of cardiovascular events, but whether the addition of ezetimibe, a nonstatin drug that reduces intestinal cholesterol absorption, can reduce the rate of cardiovascular events further is not known. METHODS We conducted a double-blind, randomized trial involving 18,144 patients who had been hospitalized for an acute coronary syndrome within the preceding 10 days and had LDL cholesterol levels of 50 to 100 mg per deciliter (1.3 to 2.6 mmol per liter) if they were receiving lipid-lowering therapy or 50 to 125 mg per deciliter (1.3 to 3.2 mmol per liter) if they were not receiving lipid-lowering therapy. The combination of simvastatin (40 mg) and ezetimibe (10 mg) (simvastatin-ezetimibe) was compared with simvastatin (40 mg) and placebo (simvastatin monotherapy). The primary end point was a composite of cardiovascular death, nonfatal myocardial infarction, unstable angina requiring rehospitalization, coronary revascularization (≥30 days after randomization), or nonfatal stroke. The median follow-up was 6 years. RESULTS The median time-weighted average LDL cholesterol level during the study was 53.7 mg per deciliter (1.4 mmol per liter) in the simvastatin-ezetimibe group, as compared with 69.5 mg per deciliter (1.8 mmol per liter) in the simvastatin-monotherapy group (P<0.001). The Kaplan-Meier event rate for the primary end point at 7 years was 32.7% in the simvastatin-ezetimibe group, as compared with 34.7% in the simvastatin-monotherapy group (absolute risk difference, 2.0 percentage points; hazard ratio, 0.936; 95% confidence interval, 0.89 to 0.99; P=0.016). Rates of prespecified muscle, gallbladder, and hepatic adverse effects and cancer were similar in the two groups. CONCLUSIONS When added to statin therapy, ezetimibe resulted in incremental lowering of LDL cholesterol levels and improved cardiovascular outcomes. Moreover, lowering LDL cholesterol to levels below previous targets provided additional benefit. (Funded by Merck; IMPROVE-IT ClinicalTrials.gov number, NCT00202878.).


European Heart Journal | 2003

Management of acute myocardial infarction in patients presenting with ST-segment elevation

Frans Van de Werf; Diego Ardissino; Amadeo Betriu; Dennis V. Cokkinos; Erling Falk; Keith A.A. Fox; Desmond G. Julian; Maria Lengyel; Franz-Josef Neumann; Witold Rużyłło; Christian Thygesen; S. Richard Underwood; Alec Vahanian; Freek W.A. Verheugt; William Wijns

Summary: initial diagnosis of acute myocar-dial infarction •History of chest pain/discomfort.•ST-segment elevations or (presumed) new leftbundle-branch block on admission ECG. RepeatedECG recordings often needed.•Elevated markers of myocardial necrosis (CK-MB,troponins). One should not wait for the results toinitiate reperfusion treatment!•2D echocardiography and perfusion scintigraphyhelpful to rule out acute myocardial infarction. Relief of pain, breathlessness and anxiety Relief of pain is of paramount importance, not onlyfor humane reasons but because the pain is associ-ated with sympathetic activation which causesvasoconstriction and increases the workload of theheart. Intravenous opioids—morphine or, whereavailable, diamorphine—are the analgesics mostcommonly used in this context (e.g. 4 to 8 mgmorphine with additional doses of 2 mg at intervalsof 5 min until the pain is relieved); intramuscularinjections should be avoided. Repeated dosesmay be necessary. Side effects include nausea andvomiting, hypotension with bradycardia, andrespiratory depression. Antiemetics may be admin-istered concurrently with opioids. The hypotensionand bradycardia will usually respond to atropine,and respiratory depression to naloxone, whichshould always be available. If opioids fail to relievethe pain after repeated administration, intra-venous beta-blockers or nitrates are sometimeseffective. Oxygen (2– 4l.min


The New England Journal of Medicine | 2011

Apixaban with Antiplatelet Therapy after Acute Coronary Syndrome

John H. Alexander; Renato D. Lopes; Stefan James; Rakhi Kilaru; Yaohua He; Puneet Mohan; Deepak L. Bhatt; Shaun G. Goodman; Freek W.A. Verheugt; Marcus Flather; Kurt Huber; Danny Liaw; Steen Husted; Jose Lopez-Sendon; Raffaele De Caterina; Petr Jansky; Harald Darius; Dragos Vinereanu; Jan H. Cornel; Frank Cools; Dan Atar; Jose Luis Leiva-Pons; Matyas Keltai; Hisao Ogawa; Prem Pais; Alexander Parkhomenko; Witold Rużyłło; Rafael Diaz; Harvey D. White; Mikhail Ruda

BACKGROUND Apixaban, an oral, direct factor Xa inhibitor, may reduce the risk of recurrent ischemic events when added to antiplatelet therapy after an acute coronary syndrome. METHODS We conducted a randomized, double-blind, placebo-controlled clinical trial comparing apixaban, at a dose of 5 mg twice daily, with placebo, in addition to standard antiplatelet therapy, in patients with a recent acute coronary syndrome and at least two additional risk factors for recurrent ischemic events. RESULTS The trial was terminated prematurely after recruitment of 7392 patients because of an increase in major bleeding events with apixaban in the absence of a counterbalancing reduction in recurrent ischemic events. With a median follow-up of 241 days, the primary outcome of cardiovascular death, myocardial infarction, or ischemic stroke occurred in 279 of the 3705 patients (7.5%) assigned to apixaban (13.2 events per 100 patient-years) and in 293 of the 3687 patients (7.9%) assigned to placebo (14.0 events per 100 patient-years) (hazard ratio with apixaban, 0.95; 95% confidence interval [CI], 0.80 to 1.11; P=0.51). The primary safety outcome of major bleeding according to the Thrombolysis in Myocardial Infarction (TIMI) definition occurred in 46 of the 3673 patients (1.3%) who received at least one dose of apixaban (2.4 events per 100 patient-years) and in 18 of the 3642 patients (0.5%) who received at least one dose of placebo (0.9 events per 100 patient-years) (hazard ratio with apixaban, 2.59; 95% CI, 1.50 to 4.46; P=0.001). A greater number of intracranial and fatal bleeding events occurred with apixaban than with placebo. CONCLUSIONS The addition of apixaban, at a dose of 5 mg twice daily, to antiplatelet therapy in high-risk patients after an acute coronary syndrome increased the number of major bleeding events without a significant reduction in recurrent ischemic events. (Funded by Bristol-Myers Squibb and Pfizer; APPRAISE-2 ClinicalTrials.gov number, NCT00831441.).


The New England Journal of Medicine | 2012

Prasugrel versus Clopidogrel for Acute Coronary Syndromes without Revascularization

Matthew T. Roe; Paul W. Armstrong; Keith A.A. Fox; Harvey D. White; Dorairaj Prabhakaran; Shaun G. Goodman; Jan H. Cornel; Deepak L. Bhatt; Peter Clemmensen; Felipe Martinez; Diego Ardissino; José Carlos Nicolau; William E. Boden; Paul A. Gurbel; Witold Rużyłło; Anthony J. Dalby; Darren K. McGuire; Jose Luis Leiva-Pons; Alexander Parkhomenko; Shmuel Gottlieb; Gracita O. Topacio; Christian W. Hamm; Gregory Pavlides; Assen Goudev; Ali Oto; Chuen Den Tseng; Béla Merkely; Vladimir Gašparović; Ramón Corbalán; Mircea Cintezǎ

BACKGROUND The effect of intensified platelet inhibition for patients with unstable angina or myocardial infarction without ST-segment elevation who do not undergo revascularization has not been delineated. METHODS In this double-blind, randomized trial, in a primary analysis involving 7243 patients under the age of 75 years receiving aspirin, we evaluated up to 30 months of treatment with prasugrel (10 mg daily) versus clopidogrel (75 mg daily). In a secondary analysis involving 2083 patients 75 years of age or older, we evaluated 5 mg of prasugrel versus 75 mg of clopidogrel. RESULTS At a median follow-up of 17 months, the primary end point of death from cardiovascular causes, myocardial infarction, or stroke among patients under the age of 75 years occurred in 13.9% of the prasugrel group and 16.0% of the clopidogrel group (hazard ratio in the prasugrel group, 0.91; 95% confidence interval [CI], 0.79 to 1.05; P=0.21). Similar results were observed in the overall population. The prespecified analysis of multiple recurrent ischemic events (all components of the primary end point) suggested a lower risk for prasugrel among patients under the age of 75 years (hazard ratio, 0.85; 95% CI, 0.72 to 1.00; P=0.04). Rates of severe and intracranial bleeding were similar in the two groups in all age groups. There was no significant between-group difference in the frequency of nonhemorrhagic serious adverse events, except for a higher frequency of heart failure in the clopidogrel group. CONCLUSIONS Among patients with unstable angina or myocardial infarction without ST-segment elevation, prasugrel did not significantly reduce the frequency of the primary end point, as compared with clopidogrel, and similar risks of bleeding were observed. (Funded by Eli Lilly and Daiichi Sankyo; TRILOGY ACS ClinicalTrials.gov number, NCT00699998.).


The New England Journal of Medicine | 2012

Thrombin-Receptor Antagonist Vorapaxar in Acute Coronary Syndromes

Pierluigi Tricoci; Zhen Huang; Claes Held; David J. Moliterno; Paul W. Armstrong; Frans Van de Werf; Harvey D. White; Philip E. Aylward; Lars Wallentin; Edmond Chen; Yuliya Lokhnygina; Jinglan Pei; Sergio Leonardi; Tyrus Rorick; A. Kilian; Lisa K. Jennings; Giuseppe Ambrosio; Christoph Bode; Angel Cequier; Jan H. Cornel; Rafael Diaz; Aycan Fahri Erkan; Kurt Huber; Michael P. Hudson; Lixin Jiang; J. Wouter Jukema; Basil S. Lewis; A. Michael Lincoff; Gilles Montalescot; José Carlos Nicolau

BACKGROUND Vorapaxar is a new oral protease-activated-receptor 1 (PAR-1) antagonist that inhibits thrombin-induced platelet activation. METHODS In this multinational, double-blind, randomized trial, we compared vorapaxar with placebo in 12,944 patients who had acute coronary syndromes without ST-segment elevation. The primary end point was a composite of death from cardiovascular causes, myocardial infarction, stroke, recurrent ischemia with rehospitalization, or urgent coronary revascularization. RESULTS Follow-up in the trial was terminated early after a safety review. After a median follow-up of 502 days (interquartile range, 349 to 667), the primary end point occurred in 1031 of 6473 patients receiving vorapaxar versus 1102 of 6471 patients receiving placebo (Kaplan-Meier 2-year rate, 18.5% vs. 19.9%; hazard ratio, 0.92; 95% confidence interval [CI], 0.85 to 1.01; P=0.07). A composite of death from cardiovascular causes, myocardial infarction, or stroke occurred in 822 patients in the vorapaxar group versus 910 in the placebo group (14.7% and 16.4%, respectively; hazard ratio, 0.89; 95% CI, 0.81 to 0.98; P=0.02). Rates of moderate and severe bleeding were 7.2% in the vorapaxar group and 5.2% in the placebo group (hazard ratio, 1.35; 95% CI, 1.16 to 1.58; P<0.001). Intracranial hemorrhage rates were 1.1% and 0.2%, respectively (hazard ratio, 3.39; 95% CI, 1.78 to 6.45; P<0.001). Rates of nonhemorrhagic adverse events were similar in the two groups. CONCLUSIONS In patients with acute coronary syndromes, the addition of vorapaxar to standard therapy did not significantly reduce the primary composite end point but significantly increased the risk of major bleeding, including intracranial hemorrhage. (Funded by Merck; TRACER ClinicalTrials.gov number, NCT00527943.).


European Heart Journal | 2009

Intracoronary infusion of bone marrow-derived selected CD34+CXCR4+ cells and non-selected mononuclear cells in patients with acute STEMI and reduced left ventricular ejection fraction: results of randomized, multicentre Myocardial Regeneration by Intracoronary Infusion of Selected Population of Stem Cells in Acute Myocardial Infarction (REGENT) Trial

Michal Tendera; Wojciech Wojakowski; Witold Rużyłło; Lidia Chojnowska; Cezary Kępka; Wiesława Tracz; Piotr Musialek; Wiesława Piwowarska; Jadwiga Nessler; Pawel Buszman; Stefan Grajek; Piotr Bręborowicz; Marcin Majka; Mariusz Z. Ratajczak

AIMS Comparison of intracoronary infusion of bone marrow (BM)-derived unselected mononuclear cells (UNSEL) and selected CD34(+)CXCR4(+) cells (SEL) in patients with acute myocardial infarction (AMI) and reduced <40% left ventricular ejection fraction (LVEF). METHODS AND RESULTS Two hundred patients were randomized to intracoronary infusion of UNSEL (n = 80) or SEL (n = 80) BM cells or to the control (CTRL) group without BM cell treatment. Primary endpoint: change of LVEF and volumes measured by magnetic resonance imaging before and 6 months after the procedure. After 6 months, LVEF increased by 3% (P = 0.01) in patients treated with UNSEL, 3% in patients receiving SEL (P = 0.04) and remained unchanged in CTRL group (P = 0.73). There were no significant differences in absolute changes of LVEF between the groups. Absolute changes of left ventricular end-systolic volume and left ventricular end-diastolic volume were not significantly different in all groups. Significant increase of LVEF was observed only in patients treated with BM cells who had baseline LVEF < median (37%). Baseline LVEF < median and time from the onset of symptoms to primary percutaneous coronary intervention > or = median were predictors of LVEF improvement in patients receiving BM cells. There were no differences in major cardiovascular event (death, re-infarction, stroke, target vessel revascularization) between groups. CONCLUSION In patients with AMI and impaired LVEF, treatment with BM cells does not lead to a significant improvement of LVEF or volumes. There was however a trend in favour of cell therapy in patients with most severely impaired LVEF and longer delay between the symptoms and revascularization.


Circulation | 2009

Apixaban, an oral, direct, selective factor Xa inhibitor, in combination with antiplatelet therapy after acute coronary syndrome: results of the Apixaban for Prevention of Acute Ischemic and Safety Events (APPRAISE) trial

John H. Alexander; Richard C. Becker; Deepak L. Bhatt; Frank Cools; Filippo Crea; Mikael Dellborg; Keith A.A. Fox; Shaun G. Goodman; Robert A. Harrington; Kurt Huber; Steen Husted; Basil S. Lewis; Jose Lopez-Sendon; Puneet Mohan; Gilles Montalescot; Mikhail Ruda; Witold Rużyłło; Freek W.A. Verheugt; Lars Wallentin

Background— After an acute coronary syndrome, patients remain at risk of recurrent events. Apixaban, an oral direct factor Xa inhibitor, is a novel anticoagulant that may reduce these events but also poses a risk of bleeding. Methods and Results— Apixaban for Prevention of Acute Ischemic and Safety Events (APPRAISE) was a phase 2, double-blind, placebo-controlled, dose-ranging study. Patients (n=1715) with recent ST-elevation or non–ST-elevation acute coronary syndrome were randomized to 6 months of placebo (n=611) or 1 of 4 doses of apixaban: 2.5 mg twice daily (n=317), 10 mg once daily (n=318), 10 mg twice daily (n=248), or 20 mg once daily (n=221). Nearly all patients received aspirin; 76% received clopidogrel. The primary outcome was International Society of Thrombosis and Hemostasis major or clinically relevant nonmajor bleeding. A secondary outcome was cardiovascular death, myocardial infarction, severe recurrent ischemia, or ischemic stroke. At the recommendation of the Data Monitoring Committee, the 2 higher-dose apixaban arms were discontinued because of excess total bleeding. Compared with placebo, apixaban 2.5 mg twice daily (hazard ratio, 1.78; 95% confidence interval, 0.91 to 3.48; P=0.09) and 10 mg once daily (hazard ratio, 2.45; 95% confidence interval, 1.31 to 4.61; P=0.005) resulted in a dose-dependent increase in major or clinically relevant nonmajor bleeding. Apixaban 2.5 mg twice daily (hazard ratio, 0.73; 95% confidence interval, 0.44 to 1.19; P=0.21) and 10 mg once daily (hazard ratio, 0.61; 95% confidence interval, 0.35 to 1.04; P=0.07) resulted in lower rates of ischemic events compared with placebo. The increase in bleeding was more pronounced and the reduction in ischemic events was less evident in patients taking aspirin plus clopidogrel than in those taking aspirin alone. Conclusions— We observed a dose-related increase in bleeding and a trend toward a reduction in ischemic events with the addition of apixaban to antiplatelet therapy in patients with recent acute coronary syndrome. The safety and efficacy of apixaban may vary depending on background antiplatelet therapy. Further testing of apixaban in patients at risk of recurrent ischemic events is warranted.


JAMA | 2008

Effect of rimonabant on progression of atherosclerosis in patients with abdominal obesity and coronary artery disease : the STRADIVARIUS randomized controlled trial

Steven E. Nissen; Stephen J. Nicholls; Kathy Wolski; Josep Rodés-Cabau; Christopher P. Cannon; John Deanfield; Jean-Pierre Després; John J. P. Kastelein; Steven R. Steinhubl; Samir Kapadia; Muhammad Yasin; Witold Rużyłło; Christophe Gaudin; Bernard Job; Bo Hu; Deepak L. Bhatt; A. Michael Lincoff; E. Murat Tuzcu

CONTEXT Abdominal obesity is associated with metabolic abnormalities and increased risk of atherosclerotic cardiovascular disease. However, no obesity management strategy has demonstrated the ability to slow progression of coronary disease. OBJECTIVE To determine whether weight loss and metabolic effects of the selective cannabinoid type 1 receptor antagonist rimonabant reduces progression of coronary disease in patients with abdominal obesity and the metabolic syndrome. DESIGN, SETTING, AND PATIENTS Randomized, double-blinded, placebo-controlled, 2-group, parallel-group trial (enrollment December 2004-December 2005) comparing rimonabant with placebo in 839 patients at 112 centers in North America, Europe, and Australia. INTERVENTIONS Patients received dietary counseling, were randomized to receive rimonabant (20 mg daily) or matching placebo, and underwent coronary intravascular ultrasonography at baseline (n = 839) and study completion (n = 676). MAIN OUTCOME MEASURES The primary efficacy parameter was change in percent atheroma volume (PAV); the secondary efficacy parameter was change in normalized total atheroma volume (TAV). RESULTS In the rimonabant vs placebo groups, PAV (95% confidence interval [CI]) increased 0.25% (-0.04% to 0.54%) vs 0.51% (0.22% to 0.80%) (P = .22), respectively, and TAV decreased 2.2 mm3 (-4.09 to -0.24) vs an increase of 0.88 mm3 (-1.03 to 2.79) (P = .03). In the rimonabant vs placebo groups, imputing results based on baseline characteristics for patients not completing the trial, PAV increased 0.25% (-0.04% to 0.55%) vs 0.57% (0.29% to 0.84%) (P = .13), and TAV decreased 1.95 mm3 (-3.8 to -0.10) vs an increase of 1.19 mm3 (-0.73 to 3.12) (P = .02). Rimonabant-treated patients had a larger reduction in body weight (4.3 kg [-5.1 to -3.5] vs 0.5 kg [-1.3 to 0.3]) and greater decrease in waist circumference (4.5 cm [-5.4 to -3.7] vs 1.0 cm [-1.9 to -0.2]) (P < .001 for both comparisons). In the rimonabant vs placebo groups, high-density lipoprotein cholesterol levels increased 5.8 mg/dL (4.9 to 6.8) (22.4%) vs 1.8 mg/dL (0.9 to 2.7) (6.9%) (P < .001), and median triglyceride levels decreased 24.8 mg/dL (-35.4 to -17.3) (20.5%) vs 8.9 mg/dL (-14.2 to -1.8) (6.2%) (P < .001). Rimonabant-treated patients had greater decreases in high-sensitivity C-reactive protein (1.3 mg/dL [-1.7 to -1.2] [50.3%] vs 0.9 mg/dL [-1.4 to -0.5] [30.9%]) and less increase in glycated hemoglobin levels (0.11% [0.02% to 0.20%] vs 0.40% [0.31% to 0.49%]) (P < .001 for both comparisons). Psychiatric adverse effects were more common in the rimonabant group (43.4% vs 28.4%, P < .001). CONCLUSIONS After 18 months of treatment, the study failed to show an effect for rimonabant on disease progression for the primary end point (PAV) but showed a favorable effect on the secondary end point (TAV). Determining whether rimonabant is useful in management of coronary disease will require additional imaging and outcomes trials, which are currently under way. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00124332.


Circulation | 2006

Gender Differences in the Management and Clinical Outcome of Stable Angina

Caroline Daly; Felicity Clemens; Jose Lopez Sendon; Luigi Tavazzi; Eric Boersma; Nicholas Danchin; François Delahaye; Anselm K. Gitt; Desmond G. Julian; David Mulcahy; Witold Rużyłło; Kristian Thygesen; Freek W.A. Verheugt; Kim Fox

Background— We sought to examine the impact of gender on the investigation and subsequent management of stable angina and to assess gender differences in clinical outcome at 1 year. Methods and Results— The Euro Heart Survey of Stable Angina enrolled patients with a clinical diagnosis of stable angina on initial assessment by a cardiologist. Baseline clinical details and cardiac investigations planned or performed within a 4-week period of the assessment were recorded, and follow-up data were collected at 1 year. A total of 3779 patients were included in the survey; 42% were female. Women were less likely to undergo an exercise ECG (odds ratio, 0.81; 95% CI, 0.69 to 0.95) and less likely to be referred for coronary angiography (odds ratio, 0.59; 95% CI, 0.48 to 0.72). Antiplatelet and statin therapies were used significantly less in women than in men, both at initial assessment and at 1 year, even in those in whom coronary disease had been confirmed. Women with confirmed coronary disease were less likely to be revascularized than their male counterparts and were twice as likely to suffer death or nonfatal myocardial infarction during the 1-year follow-up period (hazard ratio, 2.09; 95% CI, 1.13 to 3.85), even after multivariable adjustment for age, abnormal ventricular function, severity of coronary disease, and diabetes. Conclusions— Significant gender bias has been identified in the use of investigations and evidence-based medical therapy in stable angina. Women were also less likely to be revascularized. The observed bias is of particular concern in light of the adverse prognosis observed among women with stable angina and confirmed coronary disease.

Collaboration


Dive into the Witold Rużyłło's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam Witkowski

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar

Mariusz Kruk

MedStar Washington Hospital Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jerzy Pręgowski

MedStar Washington Hospital Center

View shared research outputs
Top Co-Authors

Avatar

Maciej Karcz

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Łukasz A. Małek

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Piotr Hoffman

Royal Hospital for Sick Children

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge