advances in the treatment of stable coronary - emcreg.org · advances in the treatment of stable...

27
COMPLIMENTARY CME MONOGRAPH JANUARY 2018 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-INTERNATIONAL MONOGRAPH PROCEEDINGS FROM THE NOVEMBER 12, 2017 SYMPOSIUM ANAHEIM, CA

Upload: phamanh

Post on 17-Apr-2018

219 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

COMPLIMENTARY CME MONOGRAPH

www.emcreg.org

COLLABORATE | INVESTIGATE | EDUCATE

© Copyright EMCREG-International 2018

JANUARY2018

ADVANCES IN THETREATMENT OF STABLE CORONARY ARTERY DISEASEAND PERIPHERAL ARTERY DISEASEEMCREG-INTERNATIONAL MONOGRAPH PROCEEDINGS FROM THE NOVEMBER 12, 2017 SYMPOSIUMANAHEIM, CA

Page 2: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OFSTABLE CORONARY ARTERY DISEASE

AND PERIPHERAL ARTERY DISEASE

EMCREG-International MonographJanuary 2018

Editor:

W. Brian Gibler, MD President, EMCREG-International Professor of Emergency Medicine

Department of Emergency Medicine University of Cincinnati College of Medicine

Cincinnati, OH

Associate Editor:Judy M. Racadio, MD

Assistant Editor:

Amy L. Hirsch

Production & Graphics Design Manager:Todd W. Roat

Page 3: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE i

JANUARY 2018Dear Colleagues,

The Emergency Medicine Cardiac Research and Education Group (EMCREG)-International was established in 1989 as an emergency medicine cardiovascular and neurovascular organization led by experts from the United States, Canada, and across the globe. We now have Steering Committee members from the US, Canada, Australia, Belgium, Brazil, France, Netherlands, New Zealand, Japan, Singapore, Sweden, and the United Kingdom. Now in our 28th year, we remain committed to providing you with the best educational programs and enduring material pieces possible. In addition to our usual Emergency Physician audience, we now reach out to our colleagues in Cardiology, Internal Medicine, Family Medicine, Hospital Medicine, and Emergency Medicine with our EMCREG-International University of Cincinnati Office of CME accredited symposia and enduring materials.

In this EMCREG-International Monograph, Advances in the Treatment of Stable Coronary Artery Disease and Peripheral Artery Disease, you will find a detailed discussion regarding the treatment of these two critically important disease entities. This is a Proceedings Monograph based on the 2017 EMCREG-International Satellite Symposium which was held on November 12, 2017, in Anaheim during the American Heart Association Scientific Sessions. For cardiologists, internists, family physicians, hospitalists and emergency physicians, the current approach and evolution of treatment for stable coronary artery disease (CAD) and peripheral artery disease (PAD) are particularly relevant and represent a fertile area for improving care for these patients.

This Monograph is divided into four sections. The first section provides a description of the scientific basis for the current management of patients with stable coronary artery disease. For many patients, this approach uses antiplatelet monotherapy typically aspirin. The use of dual antiplatelet therapy and anticoagulant therapy has also been evaluated in these patients. The Cardiovascular Outcomes for People Using Anticoagulation Strategies (COMPASS) trial which was published in late August 2017, is described in detail. This study was terminated prematurely because of the substantial superiority of the aspirin plus low dose rivaroxaban arm in patients with stable CAD and PAD. In the second section of this Monograph, the diagnosis and treatment of PAD is discussed in depth. Antiplatelet monotherapy serves as the predominant treatment for PAD though several other antiplatelet agents have been used as monotherapy or dual antiplatelet therapy. Balancing the positive benefits of these various therapeutic combinations versus the risk of bleeding has made monotherapy with aspirin or clopidogrel a Class Ia recommendation by the American College of Cardiology/American Heart Association Guidelines for PAD. The recently published COMPASS trial demonstrated that dual therapy with aspirin and low dose rivaroxaban was superior to treatment with aspirin only for patients with PAD. In the third section of this Monograph, a detailed discussion of the clotting mechanism emphasizes the cell based nature of the contemporary understanding of thrombosis. The intersection of the protein based clotting cascade with platelets, endothelial cells, and leukocytes represents a cohesive approach to understanding how antiplatelet and anticoagulant agents can prevent pathologic clot formation associated with disease processes such as chronic CAD and PAD. Finally, the clinical and economic value of appropriate anticoagulation with a Factor Xa inhibitor such as rivaroxaban help weave together a coherent approach to understanding the complex disease processes in stable CAD and PAD.

It is our sincere hope that you will find this EMCREG-International Proceedings Monograph from our 2017 EMCREG-International Satellite Symposium during the 2017 American Heart Association Scientific Sessions on the treatment of stable CAD and PAD useful to you in your daily practice as a cardiologist, internist, family physician, hospitalist, and emergency physician. Instructions for obtaining CME from the University of Cincinnati College of Medicine, Office of Continuing Medical Education are available at the conclusion of this January 2018, EMCREG-International Monograph. Thank you very much for your interest in EMCREG-International educational initiatives and we hope you visit our website (www.emcreg.org) for future educational events and publications.

W. Brian Gibler, MDPresident, EMCREG-InternationalProfessor of Emergency MedicineUniversity of Cincinnati College of MedicineCincinnati, Ohio USA

Page 4: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE iii

CONTRIBUTING EDITOR AND AUTHORS:

Richard C. Becker, MDProfessor of MedicineChief, Division of Cardiovascular Health and DiseaseDirector and Physician-in-ChiefUniversity of Cincinnati College of MedicineCincinnati, OH

Christopher B. Granger, MD Professor of MedicineDivision of Cardiology, Department of MedicineDirector, Cardiac Care UnitDuke University Medical CenterDurham, NC

Manesh R. Patel, MD Professor of Medicine Chief, Division of Cardiology Chief, Division of Clinical Pharmacology Duke University Medical CenterDurham, NC

W. Brian Gibler, MD (Editor)President, EMCREG-InternationalDepartment of Emergency Medicine University of Cincinnati College of MedicineCincinnati, OH

Deepak L. Bhatt, MD, MPH Professor of MedicineExecutive Director of Interventional Cardiovascular ProgramsBrigham and Women’s Hospital Heart & Vascular CenterHarvard Medical SchoolBoston, MA

Manan Pareek, MD, PhD Postdoctoral Research Fellow Brigham and Women’s Hospital Heart & Vascular CenterHarvard Medical SchoolBoston, MA

Page 5: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

iv ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

EMCREG-INTERNATIONAL MEMBERS:

W. Brian Gibler, MD, PresidentUniversity of Cincinnati Cincinnati, Ohio V. Anantharaman, MD Singapore General Hospital Singapore Tom P. Aufderheide, MD Medical College of Wisconsin Milwaukee, Wisconsin Barbra Backus, MD The Hague Medical CenterThe Hague, Netherlands Roberto R. Bassan, MD Pro-Cardiaco Hospital Rio de Janeiro, Brazil Andra L. Blomkalns, MD University of Texas Southwestern Medical Center Dallas, Texas

Richard Body, MB ChB, PhDManchester University HospitalManchester, UK Gerald X. Brogan, MD Hofstra North Shore - LIJForest Hills, New York David F. M. Brown, MD Massachusetts General Hospital Boston, Massachusetts Charles B. Cairns, MD University of ArizonaTucson, Arizona Anna Marie Chang, MD Thomas Jefferson University Philadelphia, Pennsylvania Douglas M. Char, MD Washington University School of Medicine St. Louis, Missouri Sean P. Collins, MD Vanderbilt University Nashville, Tennessee Louise Cullen, MB, BSRoyal Brisbane Hospital, BrisbaneQueensland, Australia Deborah B. Diercks, MD University of Texas Southwestern Medical Center Dallas, Texas Gregory J. Fermann, MD University of Cincinnati Cincinnati, Ohio Patrick Goldstein, MDLille University HospitalLille, France

Jin H. Han, MD Vanderbilt University Medical Center Nashville, Tennessee Katherine L. Heilpern, MD Emory University School of Medicine Atlanta, Georgia Brian Hiestand, MD, MPH Wake Forest UniversityWinston Salem, North Carolina James W. Hoekstra, MD Wake Forest University Winston Salem, North Carolina Judd E. Hollander, MD Thomas Jefferson University Philadelphia, Pennsylvania Brian R. Holroyd, MD University of Alberta Hospitals Edmonton, Alberta, Canada Shingo Hori, MD Keio University Tokyo, Japan Raymond E. Jackson, MD William Beaumont Hospital Royal Oak, Michigan J. Douglas Kirk, MD U.C. Davis Medical Center Sacramento, California Fatimah Lateef, MDSingapore General HospitalSingapore Swee Han Lim, MDSingapore General HospitalSingapore Phillip D. Levy, MD Wayne State University Detroit, Michigan Christopher R. Lindsell, PhD Vanderbilt University Nashville, Tennessee Chad V. Miller, MD Wake Forest University Winston Salem, North Carolina Richard M. Nowak, MD Henry Ford Hospital Detroit, Michigan

Masatoshi Oba, MD, PhD Osaki Citizens Hospital Osaki, Japan Gunnar Öhlén, MD, PhD Karolinska University Hospital Stockholm, Sweden

Brian J. O’Neil, MD Wayne State University Detroit, Michigan Joseph P. Ornato, MD Medical College of Virginia Richmond, Virginia Arthur M. Pancioli, MD University of Cincinnati Cincinnati, Ohio W. Frank Peacock, MD Baylor College of Medicine Houston, Texas Nicolas R. Peschanski, MD Rouen University Hospital Upper-Normandy, France Charles V. Pollack, MA, MD Thomas Jefferson University Philadelphia, Pennsylvania Emanuel P. Rivers, MD, PhD Henry Ford Hospital Detroit, Michigan Francois P. Sarasin, MD Hospital Cantonal Geneva, Switzerland Harry R. Severance, MD University of Tennessee College of Medicine Chattanooga, Tennessee Corey M. Slovis, MD Vanderbilt University Nashville, Tennessee Alan B. Storrow, MD Vanderbilt University Nashville, Tennessee Richard L. Summers, MD University of Mississippi Jackson, Mississippi Benjamin Sun, MD Oregon Health & Science UniversityPortland, Oregon

Martin Than, MDChristchurch HospitalChristchurch, New Zealand James E. Weber, MD University of Michigan Ann Arbor, Michigan

Page 6: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE v

ACCREDITATION AND DESIGNATION OF CREDIT This activity has been planned and implemented in accordance with the accreditation requirements and policies of the Accreditation Council for Continuing Medical Education through the joint providership of the University of Cincinnati and EMCREG-International. The University of Cincinnati is accredited by the ACCME to provide continuing medical education for physicians. The University of Cincinnati designates this enduring material activity for a maximum of 4.0 AMA PRA Category I Credits™. Physicians should claim only the credits commensurate with the extent of their participation in the activity. The opinions expressed during this educational activity are those of the faculty and do not necessarily represent the views of the University of Cincinnati. Participants have an implied responsibility to use the newly acquired information to enhance patient outcomes and their own professional development. The University of Cincinnati College of Medicine is committed to resolving all conflicts of interest issues, which may arise as a result of prospective faculty member’s significant relationships with drug or device manufacturer(s). The University of Cincinnati College of Medicine mandate is to retain only those speakers with financial interests that can be reconciled with the goals and educational integrity of the program. In accordance with the ACCME Standards for Commercial Support the speakers for this course have been asked to disclose to participants the existence of any financial interest/and or relationship(s) (e.g. paid speaker, employee, paid consultant on a board and/or committee for a commercial company) that would potentially affect the objectivity of his/her presentation or whose products or services may be mentioned during their presentation. The following disclosures were made:

PLANNING COMMITTEE AND FACULTY DISCLOSURES:

Planning Committee Members:

W. Brian Gibler, MD: Advisory Board: AstraZeneca, Entegrion; Shareholder: MyocardioCare, Entegrion Rick Ricer, MD: No relevant relationships Susan P. Tyler: No relevant relationships Barb Forney: No relevant relationships Speakers:

Deepak L. Bhatt, MD, MPH: Grants/Research Support Recipient: Amarin, Amgen, AstraZeneca, Bristol Myers Squibb, Chiesi, Eisai, Ethicon, Forest Laboratories, Ironwood, Ischemix, Lilly, Medtronic, Pfizer, Roche, Sanofi Aventis, The Medicines CompanyRichard C. Becker, MD: Advisor or Review Panel Member: Ionis, PortolaChristopher B. Granger, MD: Grants/Research Support Recipient: Armetheon, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Daiichi Sankyo, Duke Clinical Research Institute, FDA, Glaxo SmithKline, Janssen Pharmaceuticals, Medtronic Foundation, Novartis, Pfizer; Consultant: Abbvie, Armetheon, AstraZeneca, Bayer, Boehringer Ingelheim, Boston Scientific, Bristol Myers Squibb, Daiichi Sankyo, Gilead, Glaxo SmithKline, Janssen, Medscape, Medtronic, Merck, NIH, Novartis, Pfizer, Sirtex, VerseonManan Pareek, MD, PhD: Advisory Board: AstraZeneca; Other Relationships: AstraZeneca, MedscapeManesh R. Patel, MD Grants/Research Support Recipient: AstraZeneca, Bayer, Janssen; Advisor or Review Panel Member: AstraZeneca, Bayer, Janssen

Commercial Acknowledgment: This EMCREG-International Monograph is Supported by an educational grant from Janssen Pharmaceuticals, Inc., administered by Janssen Scientific Affairs, LLC.Disclaimer: The opinions expressed during the live activity are those of the faculty and do not necessarily represent the views of the University of Cincinnati. The information is presented for the purpose of advancing the attendees’ professional development.

Off Label Disclosure: Faculty members are required to inform the audience when they are discussing off-label, unapproved uses of devices and drugs. Physicians should consult full prescribing information before using any product mentioned during this educational activity.

Learner Assurance Statement: The University of Cincinnati is committed to resolving all conflicts of interest issues that could arise as a result of prospective faculty members’ significant relationships with drug or device manufacturer(s). The University of Cincinnati is committed to retaining only those speakers with financial interests that can be reconciled with the goals and educational integrity of the CME activity.

EMCREG-International will not be liable to you or anyone else for any decision made or action taken (or not taken) by you in reliance on these materials. This document does not replace individual physician clinical judgment. Clinical judgment must guide each professional in weighing the benefits of treatment against the risk of toxicity. Doses, indications, and methods of use for products referred to in this program are not necessarily the same as indicated in the package insert and may be derived from the professional literature or other clinical courses. Consult complete prescribing information before administering.

Page 7: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE vii

TABLE OF CONTENTS:

DECREASING MAJOR ADVERSE CLINICAL EVENTS FOR PATIENTS WITH CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE: THE COMPASS TRIAL 1 Manan Pareek, MD, PhD Postdoctoral Research FellowBrigham and Women’s Hospital Heart & Vascular CenterHarvard Medical SchoolBoston, MA

Deepak L. Bhatt, MD, MPH Professor of MedicineExecutive Director of Interventional Cardiovascular ProgramsBrigham and Women’s Hospital Heart & Vascular CenterHarvard Medical SchoolBoston, MA

IMPROVING THE TREATMENT OF PERIPHERAL ARTERY DISEASE: PROVIDING INDIVIDUALIZED, INNOVATIVE, AND EFFICIENT CARE 5Manesh R. Patel, MD Professor of Medicine Chief, Division of Cardiology Chief, Division of Clinical Pharmacology Duke University Medical Center Durham, NC

FACTOR Xa MECHANISM OF ACTION - IMPACT ON CLOTTING CASCADE, INFLAMMATION AND PLATELET ACTIVATION 11 Richard C. Becker, MD Professor of Medicine Chief, Division of Cardiovascular Health and Disease Director and Physician-in-Chief University of Cincinnati College of Medicine Cincinnati, OH

CLINICAL AND ECONOMIC VALUE OF RIVAROXABAN IN CORONARY ARTERY DISEASE 15Christopher B. Granger, MD Professor of Medicine Division of Cardiology, Department of Medicine Director, Cardiac Care Unit Duke University Medical Center Durham, NC

Page 8: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 1

REDUCING MAJOR ADVERSE CLINICAL EVENTS FOR PATIENTS WITH CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE: THE COMPASS TRIAL

REDUCING MAJOR ADVERSE CLINICAL EVENTS FOR PATIENTS WITH CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE: THE COMPASS TRIAL

Manan Pareek, MD, PhDPostdoctoral Research FellowBrigham and Women’s Hospital Heart & Vascular CenterHarvard Medical School, Boston, MA

Deepak L. Bhatt, MD, MPHProfessor of MedicineExecutive Director of Interventional Cardiovascular ProgramsBrigham and Women’s Hospital Heart & Vascular CenterHarvard Medical School, Boston, MA

Objectives

1. Describe old and recent literature regarding antiplatelet therapy in patients with chronic coronary artery disease (CAD) and peripheral artery disease (PAD).

2. Summarize recent data regarding very low-dose anticoagulation in patients with CAD and PAD.

3. Describe the tradeoff between ischemic events and bleeding events in patients with CAD and PAD treated with antithrombotic therapy.

Introduction

Chronic coronary artery disease (CAD) and peripheral artery dis-ease (PAD) are common manifestations of atherosclerosis. Recent estimates suggest that 16.5 million adults in the United States have chronic CAD.1 The prevalence of PAD is lower, affecting more than five million American adults.2 While CAD is the leading cause of death worldwide, both conditions contribute significantly to loss of disability-adjusted life-years.3,4 Atherosclerosis often manifests in several vascular beds, and the distinct clinical syndromes share many common major risk factors, including older age, smok-ing, hypertension, hypercholesterolemia, and diabetes mellitus.5 Platelets play pivotal roles in the inflammatory, thrombotic, and atherosclerotic processes. Therefore, targeting various pathways to inhibit platelet activation and aggregation is essential in preventing complications of progressive atherosclerotic disease.6

Single Agent Antiplatelet Therapy

For more than three decades, platelet inhibition with the cyclooxy-genase-1 inhibitor aspirin has been a cornerstone in the treatment and prevention of cardiovascular disease.7,8 In the secondary preventive setting, aspirin reduces the risk of myocardial infarction, stroke, or death from vascular causes by an absolute 1.5%.9 How-ever, approximately one in eight patients experiences a recurrent ischemic event while on aspirin.10

The randomized Clopidogrel versus Aspirin in Patients at Risk of Ischaemic Events (CAPRIE) trial compared clopidogrel, a P2Y12 receptor antagonist, at a dose of 75 mg once daily with aspirin 325 mg once daily in 19,185 patients with a recent ischemic stroke, a recent myocardial infarction, or symptomatic PAD.11 At a mean follow up of 1.9 years, the primary composite endpoint of ischemic stroke, myocardial infarction, or vascular death was significantly lower in the clopidogrel group (relative risk reduction, 8.7%; 95% confidence interval [CI], 0.3-16.5; p=0.04). Subgroup analyses revealed a greater risk reduction in the PAD subgroup (Figure 1). Furthermore, the benefit of clopidogrel was more pronounced among subjects at high vascular risk, including those with diabetes or multiple prior ischemic events.12,13 Importantly, clopidogrel was as least as safe as aspirin.

Given the theoretical advantages of ticagrelor over clopidogrel, the Examining Use of Ticagrelor in Peripheral Artery Disease (EUCLID) investigators randomly assigned 13,885 patients with symptomatic PAD, defined as previous lower limb revascularization or an ankle-brachial index of 0.80 or less, to receive ticagrelor 90 mg twice daily or clopidogrel 75 mg once daily.14 Poor clopidogrel metaboliz-ers per cytochrome P-450 2C19 genotyping were excluded. At a median of 30 months, the primary efficacy endpoint, i.e., a com-posite of cardiovascular death, myocardial infarction, or ischemic stroke, had occurred with similar rates in the two groups (ticagrelor versus clopidogrel, hazard ratio, 1.02; 95% CI, 0.92-1.13; p=0.65). While the subset of patients with previous revascularization experi-enced more myocardial infarctions and acute limb ischemia events, there were no differences between patients treated with ticagrelor versus clopidogrel.15 More patients in the ticagrelor group experi-enced dyspnea and any bleeding, but rates of major bleeding, fatal bleeding, and intracranial bleeding were similar for the two drugs.

CAPRIE: Clopidogrel versus Aspirin in Patients at Risk of Ischaemic Events; MI: myocardial infarction; PAD: peripheral artery disease. Reprinted with permission from CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet. 1996, 348:9038:329-1339.

01FIGURE Relative Risk Reduction (95% Con�dence

Interval) for the Primary Endpoint (a Composite of Ischemic Stroke, Myocardial Infarction, or Vascular Death) in Subgroups of the CAPRIE Trial

MI

Stroke

PAD

All patients

Aspirin better Clopidogrel better

-40 -20 0 20 40

-3.7% (-22.1 to 12.0)

7.3% (-5.7 to 18.7)

23.8% (8.9 to 36.2)

8.7% (0.3 to 16.5)

Page 9: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

2 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

REDUCING MAJOR ADVERSE CLINICAL EVENTS FOR PATIENTS WITH CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE: THE COMPASS TRIAL

Dual Antiplatelet Therapy

Dual antiplatelet therapy (DAPT) comprising aspirin and a P2Y12 receptor antagonist is the best-established regimen for patients with an acute coronary syndrome (ACS) or those undergoing percutane-ous coronary intervention and stent implantation.16,17 Accordingly, the Clopidogrel for High Atherothrombotic Risk and Ischemic Stabili-zation, Management, and Avoidance (CHARISMA) trial tested the use of DAPT with clopidogrel 75 mg daily plus aspirin 75-162 mg daily versus aspirin alone in 15,603 patients with either established vascular disease (documented CAD, cerebrovascular disease, or symptomatic PAD) or multiple atherothrombotic risk factors for a median of 28 months.18 While the primary efficacy endpoint, a composite of myocardial infarction, stroke, or death from cardio-vascular causes, was not significantly reduced with DAPT (relative risk, 0.93; 95% CI, 0.83-1.05; p=0.22), there was a significant risk reduction in the subgroup of patients with established vascular disease (p=0.045 for interaction). A post hoc analysis also showed greater efficacy of DAPT versus aspirin alone among patients with prior myocardial infarction, ischemic stroke, or symptomatic PAD.19 The relative risk reduction in the PAD subgroup was consistent with that observed for patients with myocardial infarction or stroke (Figure 2). The primary safety endpoint of severe bleeding was not significantly increased with DAPT.

DAPT using ticagrelor instead of clopidogrel was tested in the Platelet Inhibition and Patient Outcomes (PLATO) trial, in which the two P2Y12 receptor antagonists were compared in patients with an ACS. Twelve months of ticagrelor was more effective than

clopidogrel in preventing vascular events and provided a reduction in vascular death, without an increase in overall major bleeding.20 Consequently, the Prevention of Cardiovascular Events in Patients with Prior Heart Attack Using Ticagrelor Compared to Placebo on a Background of Aspirin-Thrombolysis in Myocardial Infarction 54 (PEGASUS-TIMI 54) trial examined the efficacy and safety of extended DAPT in 21,162 patients with a previous myocardial infarction and additional high-risk features for ischemic events.21 Patients received ticagrelor (90 mg twice daily or 60 mg twice daily) or placebo, on top of aspirin 75-150 mg daily. At 3 years, the primary efficacy endpoint of cardiovascular death, myocardial infarction, or stroke was significantly reduced with both ticagrelor regimens as compared with placebo: ticagrelor 90 mg (hazard ratio, 0.85; 95% CI, 0.75-0.96; p=0.008) and ticagrelor 60 mg (hazard ratio, 0.84; 95% CI, 0.74-0.95; p=0.004). Absolute risk reductions with extended DAPT were greater among subjects with diabetes and those with PAD, due to their higher baseline risk of ischemic events.22 Ticagrelor also reduced the risk of acute limb ischemia or peripheral artery revascularization. The rate of major bleeding was significantly greater with ticagrelor, but rates of intracranial hemor-rhage or fatal bleeding were similar in the three groups.

An alternative DAPT strategy was tested in the Thrombin Receptor Antagonist in Secondary Prevention of Atherothrombotic Ischemic Events-TIMI 50 (TRA 2°P-TIMI 50) trial, in which 26,449 patients with a history of myocardial infarction, ischemic stroke, or symp-tomatic PAD were randomly assigned to receive either vorapaxar, a protease-activated receptor 1 antagonist, at a dose of 2.5 mg daily or placebo, on a background of antiplatelet therapy.23 Vorapaxar re-sulted in a significantly lower three-year risk of the primary efficacy endpoint, a composite of death from cardiovascular causes, myo-cardial infarction, or stroke (hazard ratio, 0.87; 95% CI, 0.80-0.94; p<0.001). However, this came at the expense of a significantly increased risk of moderate or severe bleeding, including intracranial hemorrhage. The risk of intracranial hemorrhage was particularly pronounced among patients with previous stroke and prompted the data and safety monitoring board to recommend discontinuation of study drug in this subgroup after a median of two years of follow up. In patients with PAD, vorapaxar did not reduce the primary endpoint; however, the group of patients assigned to vorapaxar ex-perienced significantly lower rates of hospitalization for acute limb ischemia and peripheral revascularization.24

Very Low-Dose Anticoagulation

Patients with atherosclerotic disease remain at high risk for recur-rent cardiovascular events despite antiplatelet therapy. Since these individuals also display an increased activation of the coagulation system, there has been an interest in examining the role of oral anticoagulation in this setting.25

In stented patients, a vitamin K antagonist does not offer superior protection to DAPT and is associated with an increased risk of

02FIGURE Hazard Ratio (95% Con�dence Interval) for

the Primary Endpoint (a Composite of Cardiovascular Death, Myocardial Infarction, or Stroke) in the Subgroup of Patients Enrolled Because of Myocardial Infarction, Ischemic Stroke, or Peripheral Artery Disease from the CHARISMA Trial

MI

Ischemic stroke

PAD

All patients

Clopidogrel + Aspirin better Aspirin better0.5 0.75 1 1.5 2

0.77% (0.61 to 0.98)

0.78% (0.62 to 0.98)

0.87% (0.67 to 1.13)

0.83% (0.72 to 0.96)

CHARISMA: Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management, and Avoidance; MI: Myocardial infarction; PAD: peripheral artery disease. Reprinted with permission from Bhatt DL, Flather MD, Hacke W, Beger PB, et al. Patients With Prior Myocardial Infarction, Stroke, or Symptomatic Peripheral Arterial Disease in the CHARISMA Trial. J Amer Coll Cardiol. 2007, 49:19:1982-1988.

Page 10: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 3

REDUCING MAJOR ADVERSE CLINICAL EVENTS FOR PATIENTS WITH CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE: THE COMPASS TRIAL

bleeding.26 However, a Phase 2 safety trial in patients who had been stabilized after ACS suggested that the direct factor Xa inhibi-tor, rivaroxaban, might decrease the risk of ischemic endpoints.27 The subsequent Anti-Xa Therapy to Lower Cardiovascular Events in Addition to Standard Therapy in Subjects with Acute Coronary Syndrome-TIMI 51 (ATLAS-ACS TIMI 51) trial tested the role of very low-dose rivaroxaban after ACS.28 A total of 15,526 patients were randomized to rivaroxaban, at a dose of either 2.5 mg or 5 mg twice daily, or placebo, in addition to DAPT with low-dose aspirin and a thienopyridine (clopidogrel or ticlopidine). At a mean of 13 months, the primary composite efficacy endpoint of death from cardiovascular causes, myocardial infarction, or stroke, was significantly reduced with both doses of rivaroxaban (hazard ratio, 0.84; 95% CI, 0.74-0.96; p=0.008), albeit at the expense of more major bleeding and intracranial hemorrhage. The 2.5 mg rivaroxa-ban dose was associated with lower bleeding rates than 5 mg, as well as decreased rates of death from cardiovascular causes and all-cause mortality compared to the 5 mg dose.

These intriguing findings for very low-dose anticoagulation finally culminated in the Cardiovascular Outcomes for People Using Anti-coagulation Strategies (COMPASS) trial, in which 27,395 individuals with stable atherosclerotic vascular disease (CAD or PAD) without an indication for DAPT or anticoagulation were randomly assigned to receive rivaroxaban 2.5 mg twice daily plus aspirin 100 mg once daily, rivaroxaban 5 mg twice daily, or aspirin 100 mg once daily.29 The trial was stopped early, after a mean follow up of 23 months, owing to a consistent benefit in favor of the combination therapy arm. Thus, the primary endpoint, a composite of cardio-vascular death, stroke, or myocardial infarction, was significantly reduced with rivaroxaban plus aspirin versus aspirin alone (hazard ratio, 0.76; 95% CI, 0.66-0.86; p<0.001), but not with rivaroxaban alone versus aspirin alone (hazard ratio, 0.90; 95% CI, 0.79-1.03; p=0.12). Rivaroxaban-aspirin significantly lowered the individual endpoints of cardiovascular death, death from any cause, and stroke (Figure 3). In 7,470 participants included with PAD, major limb events were substantially lowered as well (Figure 4). As expected, major bleeding was significantly increased with rivaroxa-ban-aspirin versus aspirin (hazard ratio, 1.70; 95% CI, 1.40-2.05; p<0.001) and with rivaroxaban versus aspirin (hazard ratio, 1.51; 95% CI, 1.25-1.84; p<0.001). However, the combination regimen did not significantly increase fatal or intracranial hemorrhage and was associated with significant net clinical benefit (hazard ratio, 0.80; 95% CI, 0.70-0.91; p<0.001).

Summary and Conclusions

Patients with stable atherosclerotic disease derive benefit from sec-ondary prevention with antiplatelet drugs. In this setting, clopidogrel is superior to aspirin. After an ACS event or percutaneous coronary intervention, DAPT with aspirin and a P2Y12 receptor antagonist is the preferred regimen. In many ACS patients at high risk for recurrent cardiovascular events and at low bleeding risk, extending DAPT beyond 12 months may be advantageous. The COMPASS trial has challenged the traditional antiplatelet-only paradigm by demonstrating a considerable ischemic benefit and, importantly, a reduction in cardiovascular mortality and all-cause mortality with very low-dose anticoagulation added to aspirin in patients with stable CAD and PAD. Still, as with any antithrombotic regimen, its use in clinical practice will require careful balancing of the risk of ischemia versus bleeding. Further analyses from the COMPASS trial are likely to identify individuals who will benefit the most from this new therapeutic approach.

03FIGURE Primary and Selected Secondary E�cacy

Endpoints in the COMPASS Trial (All Study Patients)

MI

Stroke

Cardiovascular death

Death from any cause

Rivaroxaban + aspirin better Aspirin better0.5 0.75 1 1.5

0.86% (0.70 to 1.05)

0.58% (0.44 to 0.76)

0.78% (0.64 to 0.96)

0.82% (0.71 to 0.96)

Primary compositie endpoint 0.76% (0.66 to 0.86)

Net clinical bene�t 0.80% (0.70 to 0.91)

COMPASS: Cardiovascular Outcomes for People Using Anticoagulation Strategies; MI: myocardial infarction.

04FIGURE Primary and Selected Secondary E�cacy

Endpoints in the COMPASS Trial (Subgroup with Peripheral Artery Disease)

MI

Stroke

Cardiovascular death

Primary compositie endpoint

Rivaroxaban + aspirin better Aspirin better0.5 0.75 1 2

0.76% (0.53 to 1.09)

0.54% (0.33 to 0.87)

0.82% (0.59 to 1.14)

0.72% (0.57 to 0.90)

MALE 0.54% (0.35 to 0.84)

Major amputation 0.30% (0.11 to 0.80)

Key composite endpoint

Net clinical bene�t

1.50.25

0.69% (0.56 to 0.85)

0.72% (0.59 to 0.87)

MALE: major adverse limb events; MI: myocardial infarction; PAD: peripheral artery disease. Figures 3 and 4 reprinted with permission from Eikelboom JW, Connolly SJ, Bosch J, et al. Rivaroxaban with or without aspirin in stable cardiovascular disease. N Engl J Med. 2017;Epub ahead of print.

Page 11: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

4 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

REDUCING MAJOR ADVERSE CLINICAL EVENTS FOR PATIENTS WITH CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE: THE COMPASS TRIAL

References

1. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, et al. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation. 2017;135(10):e146-e603.

2. Selvin E, Erlinger TP. Prevalence of and risk factors for peripheral arterial disease in the United States: results from the National Health and Nutrition Examination Survey, 1999-2000. Circulation. 2004;110(6):738-43.

3. GBD 2016 DALYs and HALE Collaborators. Global, regional, and national disability-adjusted life-years (DALYs) for 333 diseases and injuries and healthy life expectancy (HALE) for 195 countries and territories, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2017;390(10100):1260-344.

4. GBD 2016 Causes of Death Collaborators. Global, regional, and national age-sex specific mortality for 264 causes of death, 1980-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2017;390(10100):1151-210.

5. Bhatt DL, Steg PG, Ohman EM, Hirsch AT, Ikeda Y, Mas JL, et al. International prevalence, recognition, and treatment of cardiovascular r isk factors in outpatients with atherothrombosis. JAMA. 2006;295(2):180-9.

6. Meadows TA, Bhatt DL. Clinical aspects of platelet inhibitors and thrombus formation. Circ Res. 2007;100(9):1261-75.

7. Lewis HD, Jr., Davis JW, Archibald DG, Steinke WE, Smitherman TC, Doherty JE, 3rd, et al. Protective effects of aspirin against acute myocardial infarction and death in men with unstable angina. Results of a Veterans Administration Cooperative Study. N Engl J Med. 1983;309(7):396-403.

8. Cairns JA, Gent M, Singer J, Finnie KJ, Froggatt GM, Holder DA, et al. Aspirin, sulfinpyrazone, or both in unstable angina. Results of a Canadian multicenter trial. N Engl J Med. 1985;313(22):1369-75.

9. Antithrombotic Trialists Collaboration, Baigent C, Blackwell L, Collins R, Emberson J, Godwin J, et al. Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials. Lancet. 2009;373(9678):1849-60.

10. Antithrombotic Trialists Collaboration. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. BMJ. 2002;324(7329):71-86.

11. CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). CAPRIE Steering Committee. Lancet. 1996;348(9038):1329-39.

12. Ringleb PA, Bhatt DL, Hirsch AT, Topol EJ, Hacke W. Clopidogrel Versus Aspirin in Patients at Risk of Ischemic Events I. Benefit of clopidogrel over aspirin is amplified in patients with a history of ischemic events. Stroke. 2004;35(2):528-32.

13. Bhatt DL, Marso SP, Hirsch AT, Ringleb PA, Hacke W, Topol EJ. Amplified benefit of clopidogrel versus aspirin in patients with diabetes mellitus. Am J Cardiol. 2002;90(6):625-8.

14. Hiatt WR, Fowkes FG, Heizer G, Berger JS, Baumgartner I, Held P, et al. Ticagrelor versus Clopidogrel in Symptomatic Peripheral Artery Disease. N Engl J Med. 2017;376(1):32-40.

15. Jones WS, Baumgartner I, Hiatt WR, Heizer G, Conte MS, White CJ, et al. Ticagrelor Compared With Clopidogrel in Patients With Prior Lower Extremity Revascularization for Peripheral Artery Disease. Circulation. 2017;135(3):241-50.

16. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK, et al. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med. 2001;345(7):494-502.

17. Mehta SR, Yusuf S, Peters RJ, Bertrand ME, Lewis BS, Natarajan MK, et al. Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: the PCI-CURE study. Lancet. 2001;358(9281):527-33.

18. Bhatt DL, Fox KA, Hacke W, Berger PB, Black HR, Boden WE, et al. Clopidogrel and aspirin versus aspirin alone for the prevention of atherothrombotic events. N Engl J Med. 2006;354(16):1706-17.

19. Bhatt DL, Flather MD, Hacke W, Berger PB, Black HR, Boden WE, et al. Patients with prior myocardial infarction, stroke, or symptomatic peripheral arterial disease in the CHARISMA trial. J Am Coll Cardiol. 2007;49(19):1982-8.

20. Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361(11):1045-57.

21. Bonaca MP, Bhatt DL, Cohen M, Steg PG, Storey RF, Jensen EC, et al. Long-term use of ticagrelor in patients with prior myocardial infarction. N Engl J Med. 2015;372(19):1791-800.

22. Bhatt DL, Bonaca MP, Bansilal S, Angiolillo DJ, Cohen M, Storey RF, et al. Reduction in Ischemic Events With Ticagrelor in Diabetic Patients With Prior Myocardial Infarction in PEGASUS-TIMI 54. J Am Coll Cardiol. 2016;67(23):2732-40.

23. Morrow DA, Braunwald E, Bonaca MP, Ameriso SF, Dalby AJ, Fish MP, et al. Vorapaxar in the secondary prevention of atherothrombotic events. N Engl J Med. 2012;366(15):1404-13.

24. Bonaca MP, Scirica BM, Creager MA, Olin J, Bounameaux H, Dellborg M, et al. Vorapaxar in patients with peripheral artery disease: results from TRA°2 P-TIMI 50. Circulation. 2013;127(14):1522-9, 9e1-6.

25. Bhatt DL, Hulot JS, Moliterno DJ, Harrington RA. Antiplatelet and anticoagulation therapy for acute coronary syndromes. Circ Res. 2014;114(12):1929-43.

26. Leon MB, Baim DS, Popma JJ, Gordon PC, Cutlip DE, Ho KK, et al. A clinical trial comparing three antithrombotic-drug regimens after coronary-artery stenting. Stent Anticoagulation Restenosis Study Investigators. N Engl J Med. 1998;339(23):1665-71.

27. Mega JL, Braunwald E, Mohanavelu S, Burton P, Poulter R, Misselwitz F, et al. Rivaroxaban versus placebo in patients with acute coronary syndromes (ATLAS ACS-TIMI 46): a randomised, double-blind, phase II trial. Lancet. 2009;374(9683):29-38.

28. Mega JL, Braunwald E, Wiviott SD, Bassand JP, Bhatt DL, Bode C, et al. Rivaroxaban in patients with a recent acute coronary syndrome. N Engl J Med. 2012;366(1):9-19.

29. Eikelboom JW, Connolly SJ, Bosch J, Dagenais GR, Hart RG, Shestakovska O, et al. Rivaroxaban with or without Aspirin in Stable Cardiovascular Disease. N Engl J Med. 2017;377(14):1319-30.

Page 12: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 5

IMPROVING THE TREATMENT OF PERIPHERAL ARTERY DISEASE: PROVIDING INDIVIDUALIZED, INNOVATIVE, AND EFFICIENT CARE

IMPROVING THE TREATMENT OF PERIPHERAL ARTERY DISEASE: PROVIDING INDIVIDUALIZED, INNOVATIVE, AND EFFICIENT CARE

Manesh R. Patel, MDProfessor of Medicine Chief, Division of Cardiology Co-Director, Duke Heart Center Duke University, Durham, NC

Objectives

1. Describe the symptoms and health risks associated with periph-eral artery disease (PAD).

2. Summarize the current medical approach to treating patients with PAD.

3. Outline the significant results of recent trials of antithrombotic therapy in patients with PAD.

Introduction

Atherosclerotic peripheral artery disease (PAD) affects more than 200 million adults worldwide1 and an estimated eight million people in the United States. The prevalence of peripheral artery disease (PAD) in patients over 70 years of age or over 55 with diabetes is estimated near 30% from the Partners study (Figure 1). Lower extremity PAD is considered a manifestation of systemic atherosclerosis that affects the arteries of the lower limbs. Despite recent advances in diagnosis and treatment, 5-10% of patients with PAD have recurrent events and millions die from cardiovascular dis-ease each year.2 Many medical strategies are considered important for patients with PAD. These include smoking cessation, diabetes control, blood pressure management, exercise therapy for claudica-tion, and antithrombotic medications. Antithrombotic medications have been proven to reduce cardiovascular morbidity and mortal-ity in a number of scenarios, including acute coronary syndrome, atrial fibrillation, and percutaneous coronary intervention.3-7 Statin therapy is considered a cornerstone treatment to reduce the occur-rence of major adverse cardiovascular events in patients with stable atherosclerotic disease.8,9 The evidence base for PAD therapies has evolved recently. The cardiovascular risk of patients with PAD, risk reduction strategies, recent antithrombotic trial data, and opportuni-ties for care improvement moving forward will be reviewed here.

PAD Patient Population and Treatment Opportunities

Most patients with PAD are asymptomatic, and those with symp-toms can present with a variety of complaints including atypical leg pain, intermittent claudication (leg pain that occurs with exertion and improves with rest), ischemic rest pain, ulceration, or gan-grene.10 The symptom presentation often dictates how patients are identified and brought to clinical specialties. The ankle-brachial

index (ABI) is the guideline recommended and most frequently used diagnostic test to determine the presence of PAD; the degree of hemodynamic abnormality is often used along with symptoms to determine treatment strategies.

Medical treatment of patients with PAD has traditionally involved antiplatelet monotherapy (e.g., aspirin or clopidogrel) and moder-ate- to high-intensity statin medication to reduce cardiovascular risk over time.10 Although PAD is generally considered a coronary artery disease (CAD) risk equivalent, antiplatelet and statin medications are used significantly less frequently in patients with PAD than in patients with CAD. As such, there is significant opportunity to improve treatment rates and compliance with antiplatelet and statin medications in patients with PAD.11,12 In patients with persistent symptoms despite background medical therapy, cilostazol and supervised exercise training for intermittent claudication have been shown to improve walking distance and quality of life.13,14 Until recently, supervised exercise training has been seldom used by eligible patients due to lack of insurance reimbursement and sparse availability around the country. In May 2017, however, the Centers for Medicare and Medicaid Services announced a National Coverage Determination that will reimburse providers for supervised exercise training in patients with intermittent claudication.

There are few proven medical therapies for patients with critical limb ischemia (CLI), the most severe form of PAD. In patients with limb threatening ischemia, non-invasive and invasive imaging is rec-ommended to define the burden and severity of obstructive disease, and revascularization is frequently recommended to preserve limb function and mobility. Typically only 30% of patients who undergo a limb amputation have an arterial diagnostic study of any kind per-formed prior to the amputation. The heterogeneity that exists in the application of these diagnostic and interventional strategies is also

01FIGURE Age -Standardized Mortality Rate for Patients

with Peripheral Artery Disease, 2014

Reprinted with permission from Roth et al, Trends and Patterns of Geographic Variation in Cardiovascular Mortality Among US Counties, 1980-2014. JAMA 2010;317(19): 1976-1992 JAMA 2010;317(19):1976-1992.

Page 13: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

6 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

IMPROVING THE TREATMENT OF PERIPHERAL ARTERY DISEASE: PROVIDING INDIVIDUALIZED, INNOVATIVE, AND EFFICIENT CARE

geographically variable across the country.15,16 Finally, in Medicare patients the mortality rate of patients with CLI at one year is nearly 50%, signaling the need for therapies aimed at this population.17

Dual Antiplatelet Therapy

When compared with patients with other forms of atherosclerotic disease, including CAD, patients with PAD have a higher risk of cardiovascular death, myocardial infarction (MI), and stroke. In the Reduction of Atherothrombosis for Continued Health (REACH) reg-istry, PAD patients had a 21.1% annual risk of cardiovascular death, myocardial infarction, stroke, or hospitalization for an atherothrom-botic cause.18 Also, the risk of major adverse limb events, typically defined as major amputation or surgical intervention, varies from 2-10% annually depending on age, symptom classification, con-comitant medical therapy, and prior revascularization procedures.

Importantly, major amputation of the lower extremities due to PAD has decreased significantly in the United States, but it remains an important public health concern since mortality rates are nearly 50% at one year and 70% at three years after major amputation in Medicare patients.19 Lower extremity peripheral vascular interven-tions have increased significantly over the last two decades (Figure 2).

Risk Reduction from Antithrombotic Agents

Antiplatelet therapies have been the center of treatment for patients with atherosclerotic vascular disease; the American College of Cardiology/American Heart Association (ACC/AHA) guidelines place a Class Ia recommendation for antiplatelet monotherapy with aspirin (75-325 mg daily) or clopidogrel (75 mg daily) to reduce the incidence of MI, stroke, and vascular death in patients with symptomatic PAD.10 Since the data for antiplatelet therapy in

asymptomatic patients with PAD is derived from small studies and is more heterogeneous, the ACC/AHA guidelines place a Class IIa recommendation for antiplatelet therapy in these patients. There remains uncertainty about the long-term safety and efficacy of dual antiplatelet therapy in patients with PAD based on a single sub-group analysis from the Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management and Avoidance (CHARIS-MA) study, thus prolonged dual antiplatelet therapy for all patients with PAD remains a Class IIb recommendation.

There are multiple antithrombotic targets to reduce the risk of atherothrombosis in stable patients with PAD (Figure 3). Therapies have targeted platelet activity and receptors and include aspirin, clopidogrel and ticagrelor, and vorapaxar (targeting thromboxane, P2Y12, and protease-activated receptor-1 [PAR-1], respectively). Aspirin has been the dominant therapy used by vascular physicians because of its low cost, availability, and safety; however, patients remain at high risk for life threatening events such as MI and stroke despite aspirin therapy.20 With the introduction of ticlopidine21 and clopidogrel, multiple studies were performed in high-risk patients, including those with PAD. The use of ticlodipine was truncated due to an excess risk of thrombotic thrombocytopenic purpura, and while clopidogrel was found to reduce the risk of vascular death, MI, or stroke by 23.8% in the Clopidogrel versus Aspirin in Patients at Risk of Ischaemic Events (CAPRIE) trial,22 the substitution of clopi-dogrel for aspirin did not routinely occur in clinical practice due to cost. More recently, the use of oral anticoagulants has been studied in patients with atherosclerotic disease including PAD.

Recent Antithrombotic Clinical Trial Data Vorapaxar is a PAR-1 inhibitor that binds to platelets and has been studied in the setting of acute coronary syndrome and stable atherosclerotic disease (prior MI or PAD) as an addition to baseline antiplatelet therapy. In the pivotal Thrombin Receptor Antagonist in Secondary Prevention of Atherothrombotic Ischemic Events (TRA°2P)-TIMI 50 study, 26,449 patients (3,787 with PAD) were randomized to vorapaxar or placebo.23 Eighty-eight percent of these patients were receiving aspirin therapy, 37% were taking a thienopyridine, and 28% were receiving dual antiplatelet therapy on study entry. In the overall cohort, vorapaxar reduced the incidence of the composite endpoint (cardiovascular death, MI, or stroke) by 1.2%. In the PAD cohort, the risk reduction for the primary composite endpoint was not statistically significant (11.3% versus 11.9%; hazard ratio, 0.94; 95% confidence interval [CI] 0.78 – 1.14; p=0.53). Vorapaxar did reduce the risk of hospitalization for acute limb ischemia and peripheral revascularization, but the hazard of GUSTO (Global Utilization of Streptokinase and t-PA for Occluded Coronary Arteries) moderate and severe bleeding and intracranial hemorrhage was statistically significantly higher with vorapaxar. In 2014, the U.S. Food and Drug Administration approved the use of vorapaxar in patients with prior MI or PAD albeit with a warning for bleeding on the label.

02FIGURE Trends in Peripheral Vascular Intervention

(PVI) in U.S. Medicare Bene�ciaries

6.0 4.113.2 18.4 19.7

37.8

184.7174.3

186.4

215.4231.6 228.5

209.7

187.8

178.0165.3

154.8 151.6

InpatientOutpatientO�ce

2006 2007 2008 2009 2010 20110

50

100

150

200

250

Age

- and

Sex

-Adj

uste

d Ra

teof

PVI

per

100

,000

Med

icar

e Be

nefic

iari

es

Reprinted with permission from Jones WS et al, Trends in Settings of Peripheral Vascular Intervention and the Effect of Changes in the Outpatient Prospective Payment System. J Am Coll Cardiol 2015;65(9):920-7.

Page 14: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 7

IMPROVING THE TREATMENT OF PERIPHERAL ARTERY DISEASE: PROVIDING INDIVIDUALIZED, INNOVATIVE, AND EFFICIENT CARE

Another antiplatelet agent, ticagrelor, has been tested extensively in patients with PAD. The Prevention of Cardiovascular Events in Pa-tients with Prior Heart Attack Using Ticagrelor Compared to Placebo on a Background of Aspirin - Thrombolysis in Myocardial Infarction 54 (PEGASUS-TIMI 54) trial enrolled 21,162 patients with a prior history of MI, of which 1,143 had PAD.24 Patients were randomized in a 1:1:1 fashion to ticagrelor 90 mg twice daily versus ticagrelor 60 mg twice daily versus placebo on a background of aspirin. PAD patients in the ticagrelor 60 mg arm had a statistically significant reduction in cardiovascular death, MI, or stroke, but the reduction with the ticagrelor 90 mg dose was not statistically significant. Hospitalization for acute limb ischemia or peripheral revasculariza-tion was significantly reduced in the ticagrelor 90 mg arm but the reduction in the 60 mg arm was not statistically significant.24

The Examining Use of Ticagrelor in Peripheral Artery Disease (EUCLID) trial randomized 13,885 symptomatic patients with PAD in a 1:1 fashion to ticagrelor or clopidogrel monotherapy.25 Patients were followed for approximately 30 months, and there was no dif-ference between the two groups in terms of the primary composite endpoint of cardiovascular death, MI, or stroke (10.8% versus 10.6%; hazard ratio, 1.02; 95% CI, 0.92-1.13; p=0.65). Both major bleeding (1.6% versus 1.6%; hazard ratio, 1.10; 95% CI 0.84-1.43; p=0.49) and hospitalization for acute limb ischemia (1.7% versus 1.7%; hazard ratio, 1.03; 95% CI 0.79-1.33; p=0.85) were also similar between treatment groups.

03FIGURE Mechanisms of Anti-Thrombotic

Medications and Clinical Endpoints Important to Patients with PAD

Additional factors:Shear stress

In�mamation

TF + FVIIa

RivaroxabanApixaban

Dabigatran ThrombinFibrin

Platelet-Fibrin Clot Ischemic Events

Platelet Aggregation

ADP

Aspirin

TXA2

TP

P2Y12

PAR-1

Vorapaxar

Collagen / vWF

Factor Xa

Fibrinogen

+

GranuleSecretion

ClopidogrelPrasugrelTicagrelorCangrelor

Prothrombin

In the recently published Cardiovascular Outcomes for People Us-ing Anticoagulation Strategies (COMPASS) trial, a total of 27,395 patients with stable atherosclerotic vascular disease (CAD, PAD, or both) were randomized to three arms (aspirin 100 mg daily versus rivaroxaban 5 mg twice daily versus aspirin 100 mg daily plus riva-roxaban 2.5 mg twice daily) at 602 centers worldwide.26 The study was terminated earlier than expected due to overwhelming efficacy in the aspirin and low-dose rivaroxaban arm. Over approximately two years of follow up, patients randomized to aspirin plus rivaroxa-ban 2.5 mg twice daily had a significantly lower rate of the primary composite endpoint (MI, ischemic stroke, cardiovascular death) when compared with aspirin alone (4.1% versus 5.4%; hazard ratio, 0.76; 95% CI, 0.66-0.86; p<0.001). There was a significantly higher rate of major bleeding in the aspirin plus rivaroxaban group when compared with aspirin alone (3.1% versus 1.9%; hazard ratio, 1.70; 95% CI, 1.40-2.15; p<0.001). Nevertheless, there was an 18% risk reduction in all-cause mortality in favor of aspirin and low-dose rivaroxaban (3.4% versus 4.1%; hazard ratio, 0.82; 95% CI 0.71-0.96; p<0.001).

In a simultaneous report, rivaroxaban was shown to have similar ef-ficacy in the PAD cohort from the COMPASS trial. In 7,470 patients who met inclusion criteria based on a prior history of PAD, 55.2% had symptomatic limbs, 25.7% had carotid disease, and 19.1% had a low ABI. The rate of the primary composite endpoint was reduced with aspirin plus rivaroxaban 2.5 mg twice daily when compared with aspirin alone (5.1% versus 6.9%; hazard ratio, 0.72; 95% CI, 0.57-0.90; p<0.001). The risk of major bleeding was also very simi-lar to the main trial results, with aspirin plus rivaroxaban 2.5 mg twice daily being associated with a significantly higher rate of major bleeding when compared with aspirin alone (3.1% versus 1.9%; haz-ard ratio, 1.61; 95% CI, 1.12-2.31; p<0.001). However, this finding is also significant in that PAD patients did not have an elevated risk of major bleeding when compared to patients without PAD.

In aggregate, these recent trial results provide some insight into the potential pathobiology of cardiovascular and limb events in patients with PAD. These recent data demonstrated no improvement in cardiovascular outcomes with more potent mono antiplatelet therapy (EUCLID). Subgroups of studies with dual therapy versus mono antiplatelet therapy show some benefit (PEGASUS, CHA-RISMA). Finally, there is now evidence that dual pathway therapy with antiplatelet and antithrombotic therapy (COMPASS PAD) may provide the most significant cardiovascular and limb protection for PAD patients. Table 1 summarizes clinical trials of antithrom-botic agents in patients with stable peripheral arterial disease and patients undergoing peripheral revascularization.

Reprinted with permission. vWF: von Willebrand Factor; TxA2: thromboxane A2; TP: thromboxane receptor; TF: tissue factor; ADP: adenosine diphosphate.

Page 15: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

8 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

IMPROVING THE TREATMENT OF PERIPHERAL ARTERY DISEASE: PROVIDING INDIVIDUALIZED, INNOVATIVE, AND EFFICIENT CARE

01Clinical Trials of Anti-thrombotic Agents in Patients with Stable Peripheral Arterial Disease and Patients Undergoing Peripheral Revascularization

TABLE

Stable PAD 1 vs. 1 (Antiplatelet agent)

TRIALDescription

POPADADAspirin for AsymptomaticAtherosclerosis Trialists STIMS CAPRIE EUCLID

ASA 100 mg daily vs.Control1,276 patients

ASA 100 mg daily vs.Control3,350 patients

Ticlopidine vs. Placebo687 patients

ASA 100 mg daily vs.Clopidogrel6,452 patients

Ticagrelor vs. Clopidogrel13,885 patients

Majoradversecardiacevents

Vascular death, MI, stroke: 18.2% vs. 18.3%HR 0.98, 95% CI .76-1.26

Fatal or non-fatal MI/stroke or revascularization: HR 1.03, 95% CI 0.84-1.27

*All-cause mortality: 18.5% vs.26.1%

Vascular death, MI, or ischemic stroke:

HR 0.76, 95% CI 0.64-0.91

CV death, MI, or stroke: 10.8% vs. 10.6%

HR 1.02, 95% CI .92-1.13

Majoradverselimbevents

Major amputation: 2% vs. 2%

Not reported Not reported Not reported Not reported

Acutelimbischemia

Not reported Not reported Not reported Not reported 1.7% vs. 1.7%HR 1.03, 95% CI .79-1.33

Majorbleeding

Not reported HR 1.71, 95% CI 0.99-2.97 *Ticlopidine grouphad 5% rate of hemorrhagic event

Not reported for PAD subgroup

TIMI Major Bleeding:1.6% vs. 1.6%

TRIAL CHARISMA TRA2°P PEGASUS WAVE COMPASSDescription ASA vs. ASA/Clopidogrel

3,096 patientsVorapaxar vs. placebo on background of antiplatelet (88% aspirin, 28% on ASA & thienopyridine)3787 patients

Ticagrelor 90mg BID vs. Ticagrelor 60 BID vs. placebo on background of ASA1,143 patients

Antiplatelet (ASA, clopidogrel or ticlopidine) + warfarin vs. Antiplatelet2,161 patients

Rivaroxaban 2.5 mg BID + ASA 100 mg daily vs. Rivaroxaban 5 mg BID vs. ASA 100 mg daily7,470 patients

Majoradversecardiacevents

CV death, MI, or stroke:7.6% vs. 8.9%HR 0.85, 95% CI .66-1.08

CV death, MI, or stroke:11.3% vs. 11.9%HR 0.94, 95% CI .78–1.14

CV death, MI, stroke:Ticagrelor 60mg HR 0.69,95% CI .47–.99Ticagrelor 90mg HR 0.81 95% CI 0.57– 0.15

CV death, MI, or stroke:12.2% vs. 13.3%HR 0.92,95% CI 0.73-1.16

CV death, MI, or stroke:5.1% vs. 6.9%HR 0.72, 95% CI .57-.90

Majoradverselimbevents

Not reported Not reported Not reported Not reported 1.2% vs. 2.2%HR 0.54, 95% CI .35-.84

Acutelimbischemia

Not reported 2.3% vs. 3.9%HR 0.58, 95% CI 0.39–0.86

Ticagrelor 60mgHR 0.67, 95% CI .24–.87Ticagrelor 90mg HR 0.45,95% CI .14–1.45

Not reported Not reported

Majorbleeding

GUSTO Severe bleeding:1.7% vs. 1.5%HR 0.97, 95% CI .56-1.66

GUSTO Mod/Sev bleeding:7.4% vs. 4.5%HR 1.62, 95% CI 1.21–2.18

TIMI Major bleeding:Ticagrelor 60mg: HR 1.18,95% CI .29–4.70Ticagrelor 90 mg: HR 1.46,95% CI .39–5.43

Life threatening bleeding:4.0% vs. 1.2%RR 3.41, 95% CI 1.84- 6.35

ISTH Major bleeding:3.1% vs. 1.9%HR 1.61, 95% CI 1.12-2.31

Post-Revasc

TRIAL CASPAR VOYAGER ePADDescription Clopidogrel vs. placebo

on background of ASA851 patients

Rivaroxaban vs. placebo on background of ASA6,500 patients

ASA/Edoxaban vs. ASA/Clopidogrel203 patients

Majoradversecardiacevents

*Death24 patients vs. 17 HR 1.44, 95% CI .77-2.68

Not available – trial ongoing

Not reported

Majoradverselimbevents

*Primary endpoint (graft occlusion, major amputation or death)HR 0.98, 95% CI .78-1.23

Not available – trial ongoing

Not reported

Acutelimbischemia

Not reported Not available – trial ongoing

Not reported

Majorbleeding

GUSTO Severe bleeding:2.1% vs. 1.2%

Not available – trial ongoing

TIMI Major bleeding:0 patients (E) vs. 2 patients (C)

2 vs. 1 (Antiplatelet and Anticoagulant)

2 vs. 2 (Antiplatelet and Anticoagulant)

1 vs. 0 (Antiplatelet agent)

2 vs. 2 (Antiplatelet and Anticoagulant)

2 vs. 1 (Antiplatelet and Anticoagulant)

PAD: peripheral arterial dis-ease; POPADA: Prevention of Progression of Arterial Disease and Diabetes; STIMS: Swedish Ticlopidine Multicentre Study; CAPRIE: Clopidogrel versus Aspirin in Patients at Risk of Ischaemic Events; EUCLID: Examining Use of Ticagrelor in Peripheral Artery Disease; CHARISMA: Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management and Avoidance; TRA2°P: Thrombin Receptor Antagonist in Secondary Prevention of Atherothrombotic Ischemic Events; PEGASUS: Preven-tion of Cardiovascular Events in Patients with Prior Heart Attack Us-ing Ticagrelor Compared to Placebo on a Background of Aspirin; WAVE: Warfarin Antiplatelet Vascular Evaluation; COMPASS: Cardiovas-cular Outcomes for People Using Anticoagulation Strategies; CASPAR: Clopidogrel and Acetyl Salicylic Acid in Bypass Surgery for Peripheral Arterial Disease; VOYAGER: Efficacy and Safety of Rivaroxaban in Reduc-ing the Risk of Major Thrombotic Vascular Events in Subjects with Symptomatic Peripheral Artery Disease Undergoing Peripheral Revascularization Procedures of the Lower Extremity; ePAD: Edoxaban in Peripheral Arterial Disease; ASA: aspirin; MI: myocardial infarction; CV: cardiovascular; HR: hazard ratio; CI: confidence interval; RR: relative risk; TIMI: Thrombolysis in Myocardial Infarction; GUSTO: Global Utilization of Streptokinase and t-PA for Occluded Coronary Arteries; ISTH: International Society on Thrombosis and Haemostasis; BID: twice daily

Page 16: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 9

IMPROVING THE TREATMENT OF PERIPHERAL ARTERY DISEASE: PROVIDING INDIVIDUALIZED, INNOVATIVE, AND EFFICIENT CARE

Ongoing Clinical Trial

The Efficacy and Safety of Rivaroxaban in Reducing the Risk of Major Thrombotic Vascular Events in Subjects with Symptomatic Peripheral Artery Disease Undergoing Peripheral Revascularization Procedures of the Lower Extremity (VOYAGER PAD) study is a 1:1 randomized, placebo-controlled trial of rivaroxaban 2.5 mg twice daily or placebo on a background of aspirin 100 mg daily after peripheral surgical and/or endovascular revascularization. VOYAGER will enroll over 6,500 patients and should be reported in early 2019.

Conclusion

In conclusion, PAD is a systemic manifestation of atherosclerosis that affects over 200 million people worldwide. Proven therapies such as blood pressure reduction, statin therapy, and smoking cessation are variable and used less in patients with PAD compared to patients with CAD. Antithrombotic therapy for patients with PAD has recently evolved and monotherapy with clopidogrel has been shown to be similar to ticagrelor. Rivaroxaban 2.5 mg twice daily in addition to aspirin was shown to reduce cardiovascular events and limb events when compared to aspirin alone. Clinicians and pa-tients will need to have personalized discussions on how to reduce their cardiovascular and limb risk for clinical events.

References

1. Fowkes FG, Rudan D, Rudan I, Aboyans V, Denenberg JO, McDermott MM, et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: a systematic review and analysis. Lancet. 2013;382(9901):1329-40.

2. Bhatt DL, Eagle KA, Ohman EM, Hirsch AT, Goto S, et al. Comparative determinants of 4-year cardiovascular event rates in stable outpatients at risk of or with atherothrombosis. JAMA. 2010;304(12):1350-7.

3. Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357(20):2001-15.

4. Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361(11):1045-57.

5. Patel MR, Mahaffey KW, Garg J, Pan G, Singer DE, Hacke W, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365(10):883-91.

6. Granger CB, Alexander JH, McMurray JJ, Lopes RD, Hylek EM, Hanna M, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365(11):981-92.

7. Mauri L, Kereiakes DJ, Yeh RW, Driscoll-Shempp P, Cutlip DE, Steg PG, et al. Twelve or 30 months of dual antiplatelet therapy after drug-eluting stents. N Engl J Med. 2014;371(23):2155-66.

8. Heart Protection Study Collaborative G. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360(9326):7-22.

9. LaRosa JC, Grundy SM, Waters DD, Shear C, Barter P, Fruchart JC, et al. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med. 2005;352(14):1425-35.

10. Gerhard-Herman MD, Gornik HL, Barrett C, Barshes NR, Corriere MA, Drachman DE, et al. 2016 AHA/ACC Guideline on the Management of Patients With Lower Extremity Peripheral Artery Disease: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2017;69(11):1465-508.

11. Subherwal S, Patel MR, Chiswell K, Tidemann-Miller BA, Jones WS, Conte MS, et al. Clinical trials in peripheral vascular disease: pipeline and trial designs: an evaluation of the ClinicalTrials.gov database. Circulation. 2014;130(20):1812-9.

12. Armstrong EJ, Chen DC, Westin GG, Singh S, McCoach CE, Bang H, et al. Adherence to guideline-recommended therapy is associated with decreased major adverse cardiovascular events and major adverse limb events among patients with peripheral arterial disease. J Am Heart Assoc. 2014;3(2):e000697.

13. Jones WS, Schmit KM, Vemulapalli S, Subherwal S, Patel MR, Hasselblad V, et al. Treatment Strategies for Patients With Peripheral Artery Disease. Rockville (MD); 2013.

14. Vemulapalli S, Dolor RJ, Hasselblad V, Schmit K, Banks A, Heidenfelder B, et al. Supervised vs unsupervised exercise for intermittent claudication: A systematic review and meta-analysis. Am Heart J. 2015;169(6):924-37 e3.

15. Vemulapalli S, Greiner MA, Jones WS, Patel MR, Hernandez AF, Curtis LH. Peripheral arterial testing before lower extremity amputation among Medicare beneficiaries, 2000 to 2010. Circ Cardiovasc Qual Outcomes. 2014;7(1):142-50.

16. Soden PA, Zettervall SL, Curran T, Vouyouka AG, Goodney PP, Mills JL, et al. Regional variation in patient selection and treatment for lower extremity vascular disease in the Vascular Quality Initiative. J Vasc Surg. 2017;65(1):108-18.

17. Iida O, Takahara M, Soga Y, Azuma N, Nanto S, Uematsu M, et al. Prognostic Impact of Revascularization in Poor-Risk Patients With Critical Limb Ischemia: The PRIORITY Registry (Poor-Risk Patients With and Without Revascularization Therapy for Critical Limb Ischemia). JACC Cardiovasc Interv. 2017;10(11):1147-57.

18. Steg PG, Bhatt DL, Wilson PW, D'Agostino R, Sr., Ohman EM, Rother J, et al. One-year cardiovascular event rates in outpatients with atherothrombosis. JAMA. 2007;297(11):1197-206.

19. Jones WS, Patel MR, Dai D, Subherwal S, Stafford J, Calhoun S, et al. Temporal trends and geographic variation of lower-extremity amputation in patients with peripheral artery disease: results from U.S. Medicare 2000-2008. J Am Coll Cardiol. 2012;60(21):2230-6.

20. Jones WS, Patel MR, Dai D, Vemulapalli S, Subherwal S, Stafford J, et al. High mortality risks after major lower extremity amputation in Medicare patients with peripheral artery disease. Am Heart J. 2013;165(5):809-15.

21. Jones WS, Mi X, Qualls LG, Vemulapalli S, Peterson ED, Patel MR, et al. Trends in settings for peripheral vascular intervention and the effect of changes in the outpatient prospective payment system. J Am Coll Cardiol. 2015;65(9):920-7.

22. Antithrombotic Trialists Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, & stroke in high risk patients. BMJ. 2002;324(7329):71-86.

23. Janzon L. The STIMS trial: the ticlopidine experience and its clinical applications. Swedish Ticlopidine Multicenter Study. Vasc Med. 1996;1(2):141-3.

24. Bonaca MP, Bhatt DL, Storey RF, Steg PG, Cohen M, Kuder J, et al. Ticagrelor for Prevention of Ischemic Events After Myocardial Infarction in Patients With Peripheral Artery Disease. J Am Coll Cardiol. 2016;67(23):2719-28.

25. Hiatt WR, Fowkes FG, Heizer G, Berger JS, Baumgartner I, Held P, et al. Ticagrelor versus Clopidogrel in Symptomatic Peripheral Artery Disease. N Engl J Med. 2017;376(1):32-40.

26. Eikelboom JW, Connolly SJ, Bosch J, Dagenais GR, Hart RG, Shestakovska O, et al. Rivaroxaban with or without Aspirin in Stable Cardiovascular Disease. N Engl J Med. 2017.

Page 17: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 11

FACTOR Xa MECHANISM OF ACTION- IMPACT ON CLOTTING CASCADE, INFLAMMATION AND PLATELET ACTIVATION

FACTOR Xa MECHANISM OF ACTION - IMPACT ON CLOTTING CASCADE, INFLAMMATION ANDPLATELET ACTIVATION

Richard C. Becker, MD, FAHAStonehill Endowed Chair and Professor of Medicine; Chief, Division of Cardiovascular Health and Disease; Director and Physician-in-Chief, University of Cincinnati College of Medicine, Cincinnati, OH

Objectives

1. Discuss the contemporary paradigm of thrombosis, including the role of neutrophil extracellular traps (NETs), histones and DNA-histone complexes.

2. List the inflammatory and proliferative effects of Factor Xa and describe the interplay between inflammation and thrombosis.

3. Describe the evidence that attenuation of inflammation is benefi-cial in patients who are at risk for a cardiovascular event.

Introduction

A contemporary view of thrombosis emphasizes the importance of cellular surface biochemistry and the integrated contribution of platelets, leukocytes, nucleic acids, histones and perturbed endo-thelial cells. Initiation of coagulation occurs on tissue-factor (TF) bearing cells, while amplification (or priming) requires activation of platelets and coagulation proteases. The final phase, propagation, is determined by thrombin generation on platelet surfaces. The cell-based model of thrombosis highlights specific phases or biochemi-cal stages rather than a traditional view of independent coagulation pathways or cascades. Accordingly, tissue factor is considered the key element for initiation of thrombosis, wherein its ability to complex with factor VIIa (fVIIa) and activate factor X (fXa) ultimately causes thrombin generation. Although thrombin is a pivotal enzyme in thrombosis, the importance of fXa and its diverse effects on thrombin generation, inflammatory processes, smooth muscle cell proliferation and endothelial cell activation represents a point of convergence for each component part.

Factor X

Factor X (fX) is a vitamin K-dependent glycoprotein synthesized in the liver and subsequently secreted into the plasma as a precursor to an active serine protease fXa. The human protein is composed of a light chain and a heavy chain linked by a single disulfide bond. The catalytic domain of fXa is contained within the heavy chain.

Factor X is activated by excision of a small peptide from its heavy chain. The cleavage of an alanine-isoleucine peptide bond by either TF-fVIIa or fVIIIa-fIXa complex liberates the 52 amino acid peptide, providing a potentially measureable marker of fX activation. Under optimal conditions (high concentrations of TF), the TF-fVIIa complex can activate fX and, in essence, bypass the contribution of fVIII-fIX.

Prothrombinase Assembly on Platelet Surfaces

Platelets play a critical role in localizing and controlling the burst of thrombin generation leading to fibrin formation. Procoagulant phos-pholipids (microparticles), particularly phosphatidylserine, stimulate prothrombinase assembly by several orders of magnitude. Factor X activation requires a phospholipid surface; however, recent work suggests that thrombin-stimulated platelets also expose non-lipid binding sites for fVIIIa, fIXa and fXa. The platelet receptor for fXa may include membrane bound fVa, effector protease receptor (EPR-1), and an anion-exposed binding site in complex with glyco-protein Ib.

Emerging Paradigms in Thrombosis

A traditional perspective of thrombosis begins with vessel wall inju-ry and exposure of subendothelial proteins, including collagen and tissue factor, to circulating cellular and non-cellular components. Adhesion and activation of platelets, mediated by their interaction with von Willebrand protein and collagen, respectively, coupled with tissue factor-mediated activation of coagulation proteins results in thrombin generation and fibrin formation. The events as they take place on cell surfaces are summarized above. While this time-hon-ored paradigm remains firm and soundly based, emerging evidence suggests that thrombosis is much more complex and dynamic than originally believed. Several novel triggers, templates and facilita-tors, such as cell-free nucleic acids (cfNAs), histones, DNA-histone complexes, polyphosphates, and microvesicles, have recently been identified and require inclusion in the expanding universe of throm-bosis as a dominant phenotype of human conditions, disorders and diseases.

Neutrophil extracellular traps (NETs) are platforms of intact chroma-tin fibers with antimicrobial proteins that are produced by neutro-phils to ‘‘trap and disarm’’ microbes in the extracellular milieu. These NETs have been shown to interact with the vascular endothelium, platelets, red blood cells, and coagulation factors, each of which are known to participate actively in thrombus forma-tion. Specifically, NETs have been shown to induce endothelial cell death via interactions with NET-associated proteases or cationic proteins, including histones. Histones, in turn, can induce pore formation and influx of ions into cells by binding to their cellular membranes. These interactions promote increased intracellular calcium levels, endothelial activation and Weibel-Palade content release of von Willebrand factor and other prothrombotic constitu-ent proteins.

Beyond their ability to bind endothelial cells and cause activation, NETs directly activate platelets. NETs have been shown in flow systems to bind platelets and facilitate aggregation. These proper-ties are believed to be the result of both direct and indirect effects, as platelets are known to bind with histones through phospholipids, carbohydrates, and toll-like receptors (TLRs). In addition, platelets

Page 18: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

12 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

FACTOR Xa MECHANISM OF ACTION- IMPACT ON CLOTTING CASCADE, INFLAMMATION AND PLATELET ACTIVATION

can bind double-and single-stranded DNA in vitro, representing an alternative mechanism for NET-induced platelet activation.

Also, NETs may provoke thrombus formation through direct stimula-tion of both the contact and TF-mediated coagulation pathways (Figure 1). In vitro, NETs have been shown to stimulate fibrin for-mation and deposition as well as to co-localize with fibrin in blood clots. The NETs contain neutrophil elastase, which can effectively cleave TF pathway inhibitor and augment fXa activation. By binding to TF pathway inhibitor, NETs also attenuate the endothelium’s primary means to regulate TF. Last, NETs can stimulate thrombin generation and fibrin formation through fXII-mediated contact activation. Similar to cfNAs, DNA-histone complexes have prothrom-botic properties. The responsible mechanisms, however, are likely the product of inflammatory states and cellular damage rather than functional pathways.

DNA-Histone Complexes

Histones are cationic proteins that are normally found bound to DNA within the nucleus of a cell, specifically within nucleosomes. Similar to cfNAs, histones and DNA-histone complexes can be released into the circulation from dying or damaged cells. Although release of both DNA-histone complexes and NETs are hypothesized to serve primarily anti-inflammatory and pathogen restricting or constraining roles, recent studies have identified functions for these complexes in thrombosis.

Circulating histones and DNA-histone complexes have been observed in several acute and chronic inflammatory conditions. In addition, extracellular histones function as late mediators of cell damage and organ dysfunction in sepsis. Histones may provoke thrombin generation by activating platelets through stimulation of TLR2 and TLR4. In addition, histone-DNA complexes augment thrombin generation more than histones alone. Considered col-lectively, these data support the existence of an integrated and complex interface of inflammation, host-defenses and coagulation.

Factor Xa: Inflammatory and Proliferative Effects

Factor Xa binds to human umbilical vein endothelial cells via a single class of binding sites with a dissociation constant value of 6.6 ± 0.8 nM and density of 57460 ± 5200 sites per cell. The bind-ing kinetics are considered “pseudo” first order with association and dissociation constants of 0.15 × 106 m−1 s−1 and 4.0 × 10−4 s−1, respectively. FXa binding to vascular endothelial cells is not influenced by thrombin, fVa, antithrombin or TF pathway inhibitor but is blocked by antibodies specific for EPR-1, supporting its role in fXa-endothelial cell interactions. The binding of fXa is associated with the following events: 1) increased intracellular calcium; 2) increased phosphoinositide turnover; 3) tissue factor expression; 4) tissue plasminogen activator release; 5) plasminogen activator inhibitor release; 6) interleukin-6 and -8 release; 7) cellular pro-liferation; 8) expression of E-selectin, ICAM-1 (intercellular adhesion molecule) and VCAM-1 (vascular cell adhesion molecule); and 9) nitric oxide release. The ability of indirect and direct antagonists to inhibit fXa-mediated cellular effects, without impacting its surface binding capacity, suggests strongly that catalytic activity is the determining feature (Figure 2). Macrophages localized within ath-eromatous plaques can synthesize fX. An ability of fXa to promote smooth muscle cell proliferation suggests that local prothrombotic responses may also influence arterial remodeling following injury. The mitogenic response to fXa probably involves PAR-2. Functional PAR-2, an auto-activating tethered ligand, is widely distributed in human vascular endothelial cells and smooth muscle cells. FXa also exerts mitogenic effects through platelet-derived growth factor. Leukocyte proliferation has been observed following fX activation. In turn, pro-inflammatory cytokines that activate fX (fXa) are released. FXa also promotes recruitment of mast cells and their secretion of vasoactive mediators including histamine and serotonin.

01FIGURE Neutrophil Extracellular Traps (NETS) May

Provoke Thrombus Formation Through Direct Stimulation of Both The Contact and Tissue Factor-Mediated Coagulation Pathways

ROS: reactive oxygen species; RBCs: red blood cells. Reprinted with permission from Becker, RC. Aspirin and the Prevention of Venous Thromboembolism. N Engl J Med. 2012, 366; 21.

02FIGURE Integrated Cellular and Protein Clotting

Pathways Demonstrate the Importance of Cellular Involvement in This Complex Process

M: monocyte; SMC: smooth muscle cell; TF: tissue factor; NETS: neutrophil extracellular traps; MPs: mononuclear phagocytes; TLRs: toll-like receptors; PLTs: platelets; DAMPS: danger-associated molecular patterns; ROS: reactive oxygen species; LPS: lipopolysaccharide; TNF: tumor necrosis factor; PAR: proteinase activated receptors; IL: interleukin; MCP: ; VWF: von Willebrand factor; PF: platelet factor; TM: thrombomodulin; PDGF: platelet-derived growth factor; ICAM: intercellular adhesion molecule; VCAM: vascular cell adhesion molecule Reprinted with permission from Foley JH, Conway EM. Cross Talk Pathways Between Coagulation and Inflammation. Circ Res. 2016;118:1392-1408.

Page 19: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 13

FACTOR Xa MECHANISM OF ACTION- IMPACT ON CLOTTING CASCADE, INFLAMMATION AND PLATELET ACTIVATION

Translating the Anticoagulant and Anti-inflammatory Effects of Factor Xa Inhibition to Patient Care

Systemic inflammation has been implicated in coronary artery dis-ease and common phenotypes including acute coronary syndrome. Investigation of plaques points to inflammatory mechanisms as key regulators of fibrous cap fragility and the overall thrombogenic capacity of necrotic lipid core constituents. Activated macrophages, neutrophils and monocytes elaborate enzymes that degrade extracellular matrix proteins that, in turn, pave the way for plaque instability and rupture.

Clinical trials performed over the past decade suggest strongly that attenuating inflammation exerts a beneficial effect among patients at risk for coronary artery disease-related events. In addition, the re-cently completed, presented and published CANTOS trial, in which over 10,000 patients with prior myocardial infarction and elevated high-sensitivity C-reactive protein level received a monoclonal antibody targeting interleukin-1β or placebo in addition to evidence-based therapy, supports the inflammatory hypothesis of coronary artery disease and its natural history.11

The COMPASS trial was a randomized, double-blind study of 27,395 patients with stable atherosclerotic vascular disease who received either rivaroxaban, a direct inhibitor of the pleuri-potent co-agulation protease fXa, at a dose of 2.5 mg twice daily plus aspirin 100 mg daily, rivaroxaban 5 mg twice daily, or aspirin 100 mg daily. The primary outcome measure was a composite of cardiovascular death, stroke or myocardial infarction. The study was stopped for superiority of the rivaroxaban plus aspirin group after a mean follow up of 23 months.

Summary

Factor Xa is a coagulation protease that has procoagulant, proin-flammatory and proliferative effects. Its importance in atheroscle-rotic vascular disease is based on these properties that are known to underlie the pathobiology of atherosclerotic plaque development, rupture and thrombosis as the causal underpinnings for the transi-tion from stable to unstable disease and resulting clinical events. The findings from COMPASS support the importance of factor Xa and its inhibition as a viable, readily available and safe therapeutic strategy for patients with atherosclerotic vascular disease at risk for cardiovascular death, stroke and myocardial infarction.

References

1. Muller MP, Wang Y, Morrissey JH, Tajkhorshid E. Lipid specificity ofthe membrane binding domain of coagulation factor X. J ThrombHaemost. 2017;15(10):2005-16.

2. Nehaj F, Sokol J, Ivankova J, Mokan M, Kovar F, Stasko J, et al.First Evidence: TRAP-Induced Platelet Aggregation Is Reducedin Patients Receiving Xabans. Clin Appl Thromb Hemost.2017:1076029617734310.

3. Qi H, Yang S, Zhang L. Neutrophil Extracellular Traps and EndothelialDysfunction in Atherosclerosis and Thrombosis. Front Immunol.2017;8:928.

4. Helseth R, Solheim S, Arnesen H, Seljeflot I, Opstad TB. The TimeCourse of Markers of Neutrophil Extracellular Traps in PatientsUndergoing Revascularisation for Acute Myocardial Infarction or StableAngina Pectoris. Mediators Inflamm. 2016;2016:2182358.

5. Koklic T, Chattopadhyay R, Majumder R, Lentz BR. Factor Xadimerization competes with prothrombinase complex formation onplatelet-like membrane surfaces. Biochem J. 2015;467(1):37-46.

6. Eikelboom JW, Connolly SJ, Bosch J, Dagenais GR, Hart RG,Shestakovska O, et al. Rivaroxaban with or without Aspirin in StableCardiovascular Disease. N Engl J Med. 2017;377(14):1319-30.

7. Granger V, Faille D, Marani V, Noel B, Gallais Y, Szely N, et al. Humanblood monocytes are able to form extracellular traps. J Leukoc Biol.2017;102(3):775-81.

8. Kovalenko TA, Panteleev MA, Sveshnikova AN. Substrate deliverymechanism and the role of membrane curvature in factor X activationby extrinsic tenase. J Theor Biol. 2017;435:125-33.

9. Wisler JW, Becker RC. Antithrombotic therapy: new areas to understand efficacy and bleeding. Expert Opin Ther Targets. 2014;18(12):1427-34.

10. Wisler JW, Becker RC. Oral factor Xa inhibitors for the long-termmanagement of ACS. Nat Rev Cardiol. 2012;9(7):392-401.

11. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH,Ballantyne C, et al, for the CANTOS Trial Group. AntiinflammatoryTherapy with Canakinumab for Atherosclerotic Disease. N Engl J Med.2017;377(12):1119-31.

Page 20: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 15

CLINICAL AND ECONOMIC VALUE OF RIVAROXABAN IN CORONARY ARTERY DISEASE

CLINICAL AND ECONOMIC VALUE OF RIVAROXABAN IN CORONARY ARTERY DISEASE

Christopher B. Granger, MDProfessor of MedicineDuke Clinical Research Institute Division of CardiologyDuke University Medical Center, Durham, NC

Objectives

1. Discuss the balance between the benefits of antithrombotic therapy and the risk of increased bleeding in patients with atherosclerotic vascular disease.

2. Describe the efficacy and safety of non-vitamin K oral anticoagulants (NOACs) as compared to warfarin.

3. Discuss the significant results from the COMPASS trial of rivaroxaban plus aspirin versus aspirin alone in patients with stable coronary artery disease.

4. Describe the potential cost implications of using low-dose rivaroxaban in addition to aspirin to treat patients with atherosclerotic vascular disease.

Introduction

Coronary heart disease is the number one cause of death and disability in the world and is projected to continue to be so for the foreseeable future.1 An estimated 16.5 million Americans have coronary heart disease based on current data from the National Health and Nutrition Examination Survey (NHANES).2

The combination of control of risk factors and use of effective medical treatments cut the death rate from coronary heart disease in half over 20 years from 1980 to 2000.3 Patients with peripheral artery disease have fewer available options that improve outcomes. Thus, there remains a major need for more effective treatments for patients with peripheral vascular disease.

Oral Anticoagulation Prevents Vascular Events and Causes Bleeding

Although antiplatelet therapy has been the mainstay of antithrom-botic therapy for patients with stable vascular disease, there is strong evidence that oral anticoagulation with warfarin provides protection against myocardial infarction (MI). This benefit is coun-terbalanced by increased bleeding, and the net effect, including the effect on mortality, is neutral (Figure 1).4 A similar pattern has been seen in chronic heart failure without atrial fibrillation where warfarin reduces stroke but causes bleeding, resulting in a net neu-tral effect on mortality.5 Therefore oral anticoagulation has been shown to reduce arterial vascular events, but at a cost in bleeding that counterbalances the benefits. Because of this reduction in net clinical benefit, warfarin is not used for these patients.

In recent years, non-vitamin K antagonist oral anticoagulants (NOACs), which have the advantage of less life-threatening bleeding than warfarin, have been tested for treatment of vascular disease. The APPRAISE-2 trial found reduced rates of MI and stent thrombo-sis with apixaban in addition to dual antiplatelet therapy after acute coronary syndromes (ACS). This reduction in events was accom-panied by more bleeding, including intracranial hemorrhage.6 The ATLAS-2 investigators used a different strategy, testing low-dose ri-varoxaban in addition to dual antiplatelet therapy in 93% of patients without a history of stroke. They showed benefit that exceeded risk, with a reduction in mortality using the lower dose of rivaroxaban (2.5 mg twice daily) added to antiplatelet therapy.7 There was also a reduction in stent thrombosis with rivaroxaban added to antiplate-let therapy. This trial showed that oral Xa inhibitor therapy can provide overall benefit in patients with ACS. Benefit from oral factor IIa (thrombin) inhibition for patients with ACS is less clear. There is a modestly higher rate of MI with dabigatran than with warfarin across the randomized trials of atrial fibrillation and venous throm-boembolic disease.8 Phase II trials have suggested that targeting thrombin may provide some benefit following ACS, although these trials have not progressed to Phase III.

Rivaroxaban or Dabigatran with Clopidogrel - Safer than Warfarin Triple Therapy

Two completed trials have tested oral anticoagulation with warfarin versus NOACs - rivaroxaban in the PIONEER trial9 (Figure 2 and Table 1) and dabigatran in the RE-DUAL trial10 for stroke prevention in patients with atrial fibrillation who underwent stent placement and were also treated with P2Y12 inhibitor therapy. These trials found that NOACs with P2Y12 inhibitors (without aspirin) are safer than the combination of warfarin, aspirin and P2Y12 inhibitors.

MI: myocardial infarction; ASA: aspirin; CI: confidence interval. Reprinted with permission from Rothberg MB, Celestin C, Flore LD, Lawler E, et al. Warfarin plus Aspirin after Myocardial Infarction or the Acute Coronary Syndrome: Meta-Analysis with Estimates of Risk and Benefit. Ann Intern Med. 2005;143(4):241-25.

01FIGURE S econdar y Prevention with War far in and

Aspir in versus Aspir in Alone af ter Acute Coronar y Syndrome

Death

MI

Ischemic stroke

Major bleeding event

Minor bleeding event

0.96 (0.77–1.20)

0.56 (0.46–0.69)

0.46 (0.27–0.77)

2.48 (1.67–3.68)

2.65 (2.14–3.29)

Rate ratioWarfarin +ASA better

ASA better

0.05 1.0 5.0

Page 21: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

16 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

CLINICAL AND ECONOMIC VALUE OF RIVAROXABAN IN CORONARY ARTERY DISEASE

Rivaroxaban 15 mg daily without aspirin or 2.5 twice daily with as-pirin and dabigatran 110 mg or 150 mg twice daily appeared to be nearly equally effective at preventing thrombotic events, although the number of thrombotic events was too small to have high confi-dence in those findings. The 110 mg twice daily dose of dabigatran without aspirin had numerically more MIs and stent thromboses than warfarin with aspirin, although the differences were not statis-tically significant. The use of rivaroxaban 15 mg daily or dabigatran 150 mg twice daily with a P2Y12 inhibitor, but without aspirin be-yond the first few days after coronary stenting appears to result in comparable rates of stent thrombosis compared to “triple therapy” with warfarin, clopidogrel and aspirin. The observation that aspirin may not be required to prevent stent thrombosis in the presence of a NOAC and P2Y12 inhibitor was also seen in the GEMINI trial11 in which low dose rivaroxaban and clopidogrel had comparable rates of stent thrombosis as aspirin and clopidogrel. The AUGUSTUS trial will test, in a full factorial design, the impact of aspirin versus placebo combined with either warfarin or apixaban12 in patients with atrial fibrillation and coronary stenting and/or ACS.

Oral Anticoagulation and Coronary Disease Events in Patients with Atrial Fibrillation

A substantial portion of the populations in the clinical trials of NOACs versus warfarin for atrial fibrillation also had coronary artery disease. In the ROCKET-AF trial, 17% of the population had prior MI.13 Not surprisingly, these patients were at higher risk for ischemic events as well as for bleeding, and they were more likely to be on concomitant aspirin. Overall, the rates of ischemic events tended be lower with rivaroxaban than with warfarin, with a 14% reduction in hazard with rivaroxaban, p=0.05. The hazard ratio of myocardial infarction with rivaroxaban versus warfarin was 0.81 (95% confidence interval [CI], 0.63-1.06). These findings, as well as similar findings with apixaban and edoxaban, suggest that factor Xa inhibitors are at least as effective as warfarin at preventing coro-nary events with lower risk of life-threatening bleeding.

Rivaroxaban in Patients with Stable Coronary Disease Whether patients with coronary disease without atrial fibrillation may benefit from low dose rivaroxaban with or without aspirin, compared to aspirin alone, was tested in the COMPASS trial. Overall, 91% of the trial population had coronary artery disease; 20% of these were women. Half of those with prior MI had their infarction within five years of enrollment, and only 5% within one year. Importantly, the patients were on good background medical therapy to reduce vascular events, with 92% on lipid lowering drugs and 72% on angiotensin converting enzyme (ACE) inhibitors or angiotensin receptor blockers.

The 26% relative risk reduction in the primary outcome of cardiovascular death, MI, or stroke with low dose rivaroxaban plus aspirin versus aspirin alone in the coronary disease subgroup (p<0.0001) was similar to the effect in the overall trial (Figure 3). The hazard ratio for major bleeding was 1.66 (p<0.0001) with rivaroxaban plus aspirin versus aspirin. In the coronary disease population, the 1.3% absolute reduction in cardiovascular death, MI and stroke was counterbalanced by a 1.2% absolute increase in major bleeding. The overall impact on mortality becomes key to understanding the net effect. There was a 23% relative risk reduction in all-cause mortality in the coronary disease population (p=0.001),15 providing strong evidence of an overall benefit.

With respect to ischemic heart disease outcomes in the coronary disease population, MI was not significantly reduced (hazard ratio, 0.86; 95% CI, 0.70-1.05) perhaps related to small numbers, but the hazard of a broader ischemic heart disease composite (MI, coronary heart disease death, sudden death, resuscitated cardiac arrest, or unstable angina) was reduced by 17% (p=0.03, Table 2).15 The 46% relative risk reduction (p<0.0001) in stroke in this population, similar to in the overall trial, was the most striking effect on major clinical outcomes.

02FIGURE Clinical ly Signi�cant Bleeding Events

in the PIONEER Tr ial

30

25

20

15

10

5

00 30 60 90 180 270 360

Days

TIM

I Maj

or, T

IMI M

inor

, or B

leed

ing

Requ

irin

g M

edic

al A

tten

tion

(%)

VKA + DAPTRiva + DAPT

Riva + PGY12

26.7%

18%16.8%

Riva + P2Y12 v. VKA + DAPTHR=0.59 (95% CI: 0.47-0.76)p <0.001

Riva + DAPT v. VKA + DAPTHR=0.63 (95% CI: 0.50-0.80)p <0.001

Riva: rivaroxaban; DAPT: dual antiplatelet therapy; VKA: vitamin K antagonist; CI: confidence interval; CV: cardiovascular; HR: hazard ratio; MI: myocardial infarction. Figures 2 and 3 reprinted with permission from Gibson MC, Mehran R, Bode C, Halperin J, et al. Prevention of Bleeding in Patients with Atrial Fibrillation Undergoing PCI. N Engl J Med. 2016;375(25):2423-34.

01TABLE M ajor Adverse Cardiac Events in the

PIONEER Tr ial

Kaplan-Meier Estimates Hazard Ratio (95% CI)

Overall

12

Riva + P2Y12 vs.

VKA + DAPT Riva + DAPT vs. VKA + DAPT

Adverse CV Event

1.08 (0.69-1.68) p=0.750

0.93 (0.59-1.48) p=0.765

CV Death 1.29 (0.59-2.80) p=0.523

1.19 (0.54-2.62) p=0.664

MI 0.86 (0.46-1.59) p=0.625

0.75 (0.40-1.42) p=0.374

Stroke 1.07 (0.39-2.96) p=0.891

1.36 (0.52-3.58) p=0.530

Stent Thrombosis

1.20 (0.32-4.45) p=0.790

1.44 (0.40-5.09) p=0.574

Adverse CV Events + Stent Thrombosis

1.08 (0.69-1.68) P=0.750

0.93 (0.59-1.48 p=0.765

Riva +

41 (6.5%)

15 (2.4%)

19 (3.0%)

8 (1.3%)

5 (0.8%)

41 (6.5%)

P2Y(N=694)

Riva +DAPT

(N=704)

VKA +

36 (5.6%) 36 (6.0%)

14 (2.2%) 11 (1.9%)

17 (2.7%) 21 (3.5%)

10 (1.5%) 7 (1.2%)

6 (0.9%) 4 (0.7%)

36 (5.6%) 36 (6.0%)

DAPT(N=695)

Page 22: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 17

CLINICAL AND ECONOMIC VALUE OF RIVAROXABAN IN CORONARY ARTERY DISEASE

Cost Implications of Rivaroxaban for Patients with Stable Coronary Disease

The overall effects of rivaroxaban plus aspirin versus aspirin alone compare favorably to other commonly used treatments to improve outcome for patients with vascular disease, such as antiplatelet therapy, lipid lowering agents and blood pressure lowering agents. Preliminary data regarding the cost impact of rivaroxaban in the COMPASS trial have been presented.16 Examining direct costs of care, not including costs of the drug, rivaroxaban plus aspirin resulted in substantially lower health care costs than aspirin alone, driven largely by lower costs related to the reduction in stroke. There were larger differences favoring rivaroxaban in patients with peripheral artery or polyvascular arterial disease. Formal cost effectiveness analyses are ongoing.

Summary

Coronary heart disease continues to be the most important cause of death and disability in the United States and around the world. There is now another treatment proven to prevent vascular events in patients with stable coronary disease – low dose rivaroxaban added to aspirin -- with an even larger absolute benefit for patients who have both coronary disease and concomitant peripheral or cerebrovascular disease. The net benefit of low dose rivaroxaban with aspirin, compared to aspirin alone, is underscored by the 24% relative risk reduction in all-cause mortality.

References

1. Global Health Estimates 2015: Deaths by cause, age and sex, by country and by region, 2000-2015. Geneva: World Health Organization; 2016. (http://www.who.int/healthinfo/global_burden_disease/en/ accessed November 26, 2017).

2. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, et al. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation. 2017;135(10):e146-e603.

3. Ford ES, Ajani UA, Croft JB, Critchley JA, Labarthe DR, Kottke TE, et al. Explaining the decrease in U.S. deaths from coronary disease, 1980-2000. N Engl J Med. 2007;356(23):2388-98.

4. Rothberg MB, Celestin C, Fiore LD, Lawler E, Cook JR. Warfarin plus aspirin after myocardial infarction or the acute coronary syndrome: meta-analysis with estimates of risk and benefit. Ann Intern Med. 2005;143(4):241-50.

5. Homma S, Thompson JL, Pullicino PM, Levin B, Freudenberger RS, Teerlink JR, et al. Warfarin and aspirin in patients with heart failure and sinus rhythm. N Engl J Med. 2012;366(20):1859-69.

6. Alexander JH, Lopes RD, James S, Kilaru R, He Y, Mohan P, et al. Apixaban with antiplatelet therapy after acute coronary syndrome. N Engl J Med. 2011;365(8):699-708.

7. Mega JL, Braunwald E, Wiviott SD, Bassand JP, Bhatt DL, Bode C, et al. Rivaroxaban in patients with a recent acute coronary syndrome. N Engl J Med. 2012;366(1):9-19.

8. Douxfils J, Buckinx F, Mullier F, Minet V, Rabenda V, Reginster JY, et al. Dabigatran etexilate and risk of myocardial infarction, other cardiovascular events, major bleeding, and all-cause mortality: a systematic review and meta-analysis of randomized controlled trials. J Am Heart Assoc. 2014;3(3):e000515.

03FIGURE Primar y E�cac y and S afet y O utcome in the

Coronar y Ar ter y Disease Subgroup of the COMPASS Tr ial

Low-dose rivaroxaban + aspirin vs. aspirin aloneHR 0.74, 95% CI 0.65-0.86, P≤ 0.0001

Rivaroxaban alone vs. aspirin aloneHR 0.89, 95% CI 0.78-1.02, P=0.09

0 1 2 3

0

2

4

6

8

10

0

2

4

6

8

10

0

2

4

6

8

10

Cum

ulat

ive

inci

denc

e ris

k (%

)

Time (years)

Primary outcome: cardiovascular death, MI, stroke

Low-dose rivaroxaban + aspirinRivaroxaban alone

Aspirin alone

0 1 2 3

0

2

4

6

8

10

0

2

4

6

8

10

0

2

4

6

8

10

Cum

ulat

ive

inci

denc

e ris

k (%

)

Time (years)

Major Bleeding

Rivaroxaban alone

Aspirin alone

Low-dose rivaroxaban + aspirinRivaroxaban alone

Aspirin alone

Low-dose rivaroxaban + aspirin vs. aspirin aloneHR 1-66 (95% CI 1.23-2.03), P≤0.0001

Rivaroxaban alone vs. aspirin aloneHR 1-51 (95% CI 1.23-1.24), P≤0.0001

02TABLE Coronar y Disease O utcomes in the Coronar y

Disease Subgroup in the COMPASS Tr ial

Event

Rivaroxaban and aspirin

N=8,313

Aspirin N=8,261

Rivaroxaban and aspirin vs. aspirin

N (%)

N (%)

HR (95% CI) p

Myocardial infarction (MI) 169 (2%)

195 (2%)

0.86 (0.70-1.05) 0.15

MI or sudden cardiac death 234 (3%)

273 (3%)

0.85 (0.71-1.01) 0.065

MI, coronary heart disease death, SCD, resuscitated cardiac arrest, or unstable angina

264 (3%)

314 (4%)

0.83 (0.71-0.98) 0.028

HR: hazard ratio; CI: confidence interval. Reprinted with permission from Eikelboom JW, Connolly SJ, Dagenais B, Shestkovsak O, et al. Rivaroxaban with or without Aspirin in Stable Cardiovascular Disease. N Engl J Med 2017;377:1319-30.

HR: hazard ratio; CI: confidence interval. Reprinted with permission from Eikelboom JW, Connolly SJ, Dagenais B, Shestkovsak O, et al. Rivaroxaban with or without Aspirin in Stable Cardiovascular Disease. N Engl J Med 2017;377:1319-30.

Page 23: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

18 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

CLINICAL AND ECONOMIC VALUE OF RIVAROXABAN IN CORONARY ARTERY DISEASE

9. Gibson CM, Mehran R, Bode C, Halperin J, Verheugt FW, Wildgoose P, et al. Prevention of Bleeding in Patients with Atrial Fibrillation Undergoing PCI. N Engl J Med. 2016;375(25):2423-34.

10. Cannon CP, Bhatt DL, Oldgren J, Lip GYH, Ellis SG, Kimura T, et al. Dual Antithrombotic Therapy with Dabigatran after PCI in Atrial Fibrillation. N Engl J Med. 2017;377(16):1513-24.

11. Ohman EM, Roe MT, Steg PG, James SK, Povsic TJ, White J, et al. Clinically significant bleeding with low-dose rivaroxaban versus aspirin, in addition to P2Y12 inhibition, in acute coronary syndromes (GEMINI-ACS-1): a double-blind, multicentre, randomised trial. Lancet. 2017;389(10081):1799-808.

12. AUGUSTUS Trial: https://clinicaltrials.gov/ct2/show/NCT02415400.

13. Mahaffey KW, Stevens SR, White HD, Nessel CC, Goodman SG, Piccini JP, et al. Ischaemic cardiac outcomes in patients with atrial fibrillation treated with vitamin K antagonism or factor Xa inhibition: results from the ROCKET AF trial. Eur Heart J. 2014;35(4):233-41.

14. Eikelboom JW, Connolly SJ, Bosch J, Dagenais GR, Hart RG, Shestakovska O, et al. Rivaroxaban with or without Aspirin in Stable Cardiovascular Disease. N Engl J Med. 2017;377(14):1319-30.

15. Connolly SJ, Eikelboom JW, Bosch J, Dagenais G, Dyal L, Lanas F, et al. Rivaroxaban with or without aspirin in patients with stable coronary artery disease: an international, randomised, double-blind, placebo-controlled trial. Lancet. 2017.

16. Lamy A. Cost impact of rivaroxaban plus aspirin versus aspirin in the COMPASS trial. American Heart Association Scientific Sessions, Anaheim, CA, November 2017. http://professional.heart.org/professional/EducationMeetings/MeetingsLiveCME/ScientificSessions/UCM_497351_SS17-Late-Breaking-Clinical-Trials.jsp#compass.

Page 24: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 19

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

Based on the information presented in this monograph, please choose one correct response for each of the following questions or statements. Record your answers on the answer sheet found on the last page. To receive Category I credit, complete the post-test and record your responses on the following answer sheet and complete the evaluation. A passing grade of 80% is needed to receive credit.

Hardcopy test can be returnd by E-mail, Fax or Mail using the included return envelope. See following page for information. Please return the CME no later than January 15, 2019.

QUESTIONS:

1. All of the following statements regarding trials with dual antiplatelet therapy (DAPT) are true EXCEPT:A. The CHARISMA trial showed that DAPT with clopidogrel and

aspirin reduces the risk of the primary endpoint (myocardial infarction [MI], stroke, or death from cardiovascular causes) in patients with established vascular disease.

B. In the PLATO trial, twelve months of clopidogrel was more effective than ticagrelor in preventing vascular events and provided a reduction in vascular death.

C. In the PEGASUS-TIMI 54 trial, ticagrelor plus aspirin reduced the risk of acute limb ischemia or peripheral artery revascu-larization as compared to aspirin alone.

D. In the TRA 2°P-TIMI trial, vorapaxar resulted in a signifi-cantly lower 3-year risk of the primary endpoint (MI, stroke, or death from cardiovascular causes), but a significantly increased risk of moderate or severe bleeding.

2. Which of the following statements regarding medi-cal therapy for patients with atherosclerotic vascular disease is TRUE?A. Aspirin is superior to clopidogrel for prevention of cardiovas-

cular events in patients with stable atherosclerotic disease.B. Vitamin K antagonists are superior to DAPT for prevention of

cardiovascular events in patients with coronary stents.C. After an acute coronary syndrome event or percutaneous

coronary intervention, dual therapy with aspirin and rivaroxaban is the preferred regimen.

D. Low dose anticoagulation with rivaroxaban reduced cardio-vascular mortality and all-cause mortality when added to aspirin in patients with stable coronary artery disease and peripheral artery disease.

3. The following findings regarding the COMPASS trial are all true EXCEPT: A. The primary endpoint (cardiovascular death, MI, or stroke)

was significantly reduced in the rivaroxaban plus aspirin arm compared to aspirin alone.

B. The primary endpoint was significantly reduced in the rivar-oxaban alone arm compared to aspirin alone.

C. Major bleeding was significantly increased in the rivaroxaban plus aspirin arm compared to aspirin alone.

D. Major bleeding was significantly increased in the rivaroxaban alone arm compared to aspirin alone.

4. Which of the following is NOT used for DAPT in patients with acute coronary syndrome?A. RivaroxabanB. Ticagrelor C. AspirinD. Clopidogrel

5. Which of the following antithrombotic agents produced an increase in intracranial bleeding in patients with previous stroke that was significant enough to result in discontinuing the study drug in that group?A. Ticagrelor B. Rivaroxaban C. VorapaxarD. Clopidogrel

6. All of the following statements regarding treatment for patients with peripheral artery disease (PAD) are true EXCEPT:A. Diabetes control and smoking cessation are necessary.B. Statin medications are a mainstay of therapy.C. Supervised exercise therapy is used for patients with

persistent claudication.D. A diagnostic arterial study must be performed to define the

severity of obstructive vascular disease prior to limb amputation.

7. In the PEGASUS-TIMI 54 trial, which of the following antiplatelet regimens resulted in a significant reduction in the primary endpoint of cardiovascular death, MI, or stroke in the subgroup of patients with PAD?A. Aspirin aloneB. Ticagrelor 60 mg twice daily plus aspirinC. Ticagrelor 90 mg twice daily plus aspirinD. Ticagrelor 90 mg twice daily without aspirin

8. In the TRA°2P-TIMI 50 trial, patients with atherosclero-sis who were treated with vorapaxar experienced all of the following EXCEPT:A. Significantly increased rate of moderate to severe bleeding B. Significantly increased rate of intracranial hemorrhageC. Significantly reduced rate of cardiovascular events in

patients with PADD. Significantly reduced rate of hospitalization for acute limb

ischemia and peripheral revascularization (Continued Next page)

TEST ALSO AVAILABLE ONLINE: www.emcreg.org/testing*Immediate CME Certificate online with passing grade.

Continuing Medical Education Post-Test Answer Form and Evaluation

Page 25: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

20 ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE

9. All of the following statements regarding the cohort of patients with PAD in the COMPASS trial are true EXCEPT:A. Aspirin plus rivaroxaban 2.5 mg twice daily significantly

reduced the rate of the primary composite endpoint (car-diovascular death, MI, stroke) when compared with aspirin alone.

B. Aspirin plus rivaroxaban 2.5 mg twice daily significantly reduced the rate of limb events compared with aspirin alone.

C. Aspirin plus rivaroxaban 2.5 mg twice daily was associated with a significantly higher rate of major bleeding when com-pared with aspirin alone.

D. Patients with PAD had a significantly higher incidence of ma-jor bleeding than patients without peripheral arterial disease.

10. Which of the following statements regarding evidence from recent antithrombotic therapy trials is TRUE?A. Ticagrelor monotherapy significantly reduces the rate of

cardiovascular events in patients with PAD compared to clopidogrel monotherapy.

B. Dual antiplatelet therapy has not been shown to be benefi-cial in patients with PAD.

C. A combination of antiplatelet and anticoagulant therapy may provide the most significant cardiovascular and limb protec-tion for PAD patients.

D. Clopidogrel has similar efficacy to aspirin for reducing the risk of cardiovascular events in patients with atherosclerotic vascular disease.

11. Factor Xa:A. Is a coagulation proteinB. Functions at the convergence of the contact activation and

tissue factor-mediated pathways of coagulation. C. Can be inhibited effectively by medications given IV, subcuta-

neously or by mouth D. All of the above

12. Factor Xa plays a role in both thrombosis and inflammation.A. True B. False

13. A contemporary paradigm of thrombosis includes which of the following:A. Cell-free DNAB. HistonesC. Histone-DNA complexesD. All of the above

14. There is emerging evidence that medications that target inflammation in patients at risk for cardiovascu-lar events are effective.A. True B. False

15. Factor Xa inhibition, particularly when achieved in combination with a low-dose aspirin, reduces cardio-vascular events in patients at risk.A. True B. False

16. Over the twenty year period from 1980 to 2000, the death rate from coronary heart disease:A. Decreased by 50%B. Decreased by 25%C. Increased by 25%D. Did not change

17. All of the following statements regarding oral anticoag-ulation for patients with atherosclerotic heart disease are true EXCEPT:A. Non-vitamin K antagonist oral anticoagulants (NOACs) are

associated with less life-threatening bleeding than warfarin.B. Factor Xa inhibitors are likely at least as effective as warfarin

at preventing coronary events.C. Aspirin may not be required to prevent stent thrombosis in

the presence of a NOAC and P2Y12 inhibitor.D. Oral factor IIa inhibitors significantly reduce cardiovascular

events following acute coronary syndrome.

18. In the coronary disease population in the COMPASS trial, what was the effect on all-cause mortality noted in the rivaroxaban plus aspirin group compared to the aspirin alone group?A. 46% relative risk reductionB. 23% relative risk reductionC. 23% relative risk increaseD. No significant difference

19. In the coronary disease population in the COMPASS trial, the rate of which of the following events did NOT significantly decrease in the rivaroxaban plus aspirin group compared to the aspirin alone group?

A. Myocardial infarctionB. StrokeC. Ischemic heart disease compositeD. All-cause mortality

20. All of the following statements regarding oral antico-agulation trials in patients with vascular disease are true EXCEPT:A. The ATLAS-2 trial demonstrated a reduction in mortality

with low dose rivaroxaban (2.5 mg twice daily) added to antiplatelet therapy.

B. The PIONEER and RE-DUAL trials demonstrated that the combination of warfarin, aspirin and P2Y12 inhibitors is safer than NOACs with P2Y12 inhibitors (without aspirin).

C. The GEMINI trial demonstrated comparable rates of stent thrombosis in patients treated with rivaroxaban and clopido-grel as in those treated with aspirin and clopidogrel.

D. The COMPASS trial demonstrated a 26% relative risk reduc-tion in the primary outcome (MI, stroke, or cardiovascular death) in patients with coronary disease treated with rivaroxaban plus aspirin versus aspirin alone.

Continuing Medical Education Post-Test Answer Form and Evaluation

Page 26: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE 21

Continuing Medical Education Post-Test Answer Form and Evaluation

After you have read the monograph, carefully record your answers by circling the appropriate letter for each question on the CME ANSWER SHEET on this page and complete the evaluation questionnaire. CME expiration date January 15, 2019 Return the answer sheet to:EMCREG-InternationalDepartment of Emergency Medicine (ML 0769)231 Albert Sabin WayCincinnati, OH 45267-07691-855-678-5061 OR FAX TO: (888) 823-5435 OR EMAIL TO: [email protected]

Evaluation Questionnaire1. On a scale of 1 to 5, with 5 being highly satisfied and 1 being highly dissatis-

fied, please rate this program with respect to:      

Overall quality of material: 1 2 3 4 5

Content of monograph: 1 2 3 4 5

Other similar CME programs: 1 2 3 4 5

Course objectives were met: 1 2 3 4 5

2. What topics would be of interest to you for future CME programs?

3. Was there commercial or promotional bias in this monograph? □ YES □ NO If YES, please explain:

4. How long did it take for you to complete this monograph?

Name (Please Print Clearly): Email (Required):

Date of Birth: (Required): (mm/dd/yyyy)

E-mail and DOB required by CME office to generate CME certificates and create your identify for CME Office support.

Degree: Specialty: Academic Affiliation (if applicable): Address: City: State: Zip Code: Telephone Number: ( ) -

CME ANSWER SHEET

1. a b c d

2. a b c d

3. a b c d

4. a b c d

5. a b c d

6. a b c d

7. a b c d

8. a b c d

9. a b c d

10. a b c d

11. a b c d

12. a b

13. a b c d

14. a b

15. a b

16. a b c d

17. a b c d

18. a b c d 19 a b c d

20. a b c d

TEST ALSO AVAILABLE ONLINE: www.emcreg.org/testing*Immediate CME Certificate with passing grade online.

Dissatisfied      Satisfied

Page 27: advances In The Treatment Of Stable Coronary - emcreg.org · ADVANCES IN THE TREATMENT OF STABLE CORONARY ARTERY DISEASE AND PERIPHERAL ARTERY DISEASE EMCREG-International Monograph

COMPLIMENTARY CME MONOGRAPH

www.emcreg.org

COLLABORATE | INVESTIGATE | EDUCATE

© Copyright EMCREG-International 2018

JANUARY2018

ADVANCES IN THETREATMENT OF STABLE CORONARY ARTERY DISEASEAND PERIPHERAL ARTERY DISEASEEMCREG-INTERNATIONAL MONOGRAPH PROCEEDINGS FROM THE NOVEMBER 12, 2017 SYMPOSIUMANAHEIM, CA