Download as pdf or txt
Download as pdf or txt
You are on page 1of 218

PHARMACOTHERAPY SELF-ASSESSMENT PROGRAM

New!
PSAP Series
2022–2024
Available for BCPS recertification credits

PSAP Release Release Date Posttest Deadline ACPE Deadline

Cardiology January 18, 2022 July 15, 2022 January 18, 2025

Current Issues in Pharmacotherapy May 16, 2022 November 15, 2022 May 16, 2025

Behavioral Health September 15, 2022 March 15, 2023 September 15, 2025

Endocrinology and Nephrology January 17, 2023 July 17, 2023 January 17, 2026

Critical Care and Emergency May 15, 2023 November 15, 2023 May 15, 2026
Medicine

Pulmonary and Gastrointestinal September 15, 2023 March 15, 2024 September 15, 2026
Diseases

Infectious Diseases January 16, 2024 July 15, 2024 January 16, 2027

Hematology and Oncology May 15, 2024 November 15, 2024 May 15, 2027

Neurology and Chronic Conditions September 16, 2024 March 17, 2025 September 16, 2027

Series Editor Series Editor


Cynthia A. Sanoski, Daniel M. Witt,
Pharm.D., FCCP, Pharm.D., FCCP,
BCPS BCPS

For purchase information, visit www.accp.com/store


®
IMPORTANT INFORMATION ON THE RELEASE OF
PSAP 2022 Book 1 (Cardiology)
TESTING

BCPS test deadline: 11:59 p.m. (Central) on July 15, 2022.


ACPE test deadline: 11:59 p.m. (Central) on January 18, 2025.

Before submitting a posttest: Check the online errata for any changes or updates to this Pharmacotherapy Self-Assessment
Program release.

Tests may not be submitted more than one time. You may complete one or all available modules for credit. For information
on passing levels, assignment of credits, and credit reporting, see the Continuing Pharmacy Education and Recertification
Instructions pages preceding each module.

Important notice on BCPS recertification: Submitting a required posttest for BCPS recertification attests that you have completed
the test as an individual effort and not in collaboration with any other individual or group. Failure to complete this test as an
individual effort may jeopardize your ability to use PSAP for BCPS recertification.

BOOK FORMATS AND CONTENT

Online book: All purchasers of this PSAP release have access to the online book (interactive PDFs). To access, go to www.accp.com
and sign into your My Account page using your e-mail address and password (technical assistance is available). Scroll down to
find your book and the required posttests under My Products. The online book can be saved to the desktop or printed. The latest
version of Adobe Reader (available free) offers functionality such as highlighting or adding “sticky notes” to the text.

E-Media book: All purchasers also have access to the e-media version. Follow these instructions to load the text and self-assessment
questions in this book onto your e-reader, tablet, or Android phone.

PSAP Audio Companion: All purchasers also have access to the PSAP Audio Companion. Follow these instructions to load these
files onto an MP3 player or burn them onto an audio CD.

Print books: If you have purchased a print version of this book, it will be delivered on or near the release date to the address of
record on your ACCP account. If you have not received the print book within 1 week of the release date, contact customer service
by e-mailing accp@accp.com. NOTE: The online book may be updated after the print book goes to press. Before submitting a
posttest, please check the online errata (https://www.accp.com/media/psap/2016-2018/errata.pdf) for the presence of updates.

Hyperlinks: To facilitate further learning and research, this publication incorporates hyperlinks to websites administered by other
organizations. Internal and external hypertext links are visible as underlined text in the print book and are active in the Online
and e-Media versions of the book. NOTE: these links are to websites provided by third parties not affiliated in any way with ACCP.
ACCP assumes no liability for material downloaded from or accessed on these websites. It is the responsibility of the reader to
examine the copyright and licensing restrictions of linked pages and to secure all necessary permissions.

Abbreviations, Laboratory Values: At the start of each chapter/feature are hyperlinks to tables with selected medical abbreviations
and reference ranges for common laboratory tests. These tables can be used as a resource in reading the material and complet-
ing the self-assessment questions.

NOTE: The editors and publisher of PSAP recognize that the development of this volume of material offers many opportunities
for error. Despite our best efforts, some errors may persist into print. Drug dosage schedules are, we believe, accurate and in
accordance with current standards. Readers are advised, however, to check package inserts for the recommended dosages and
contraindications. This is especially important for new, infrequently used, and highly toxic drugs.
Director of International Programs, Associate Director of Professional Development: Wafa Y. Dahdal, Pharm.D.
Associate Director, Professional Development and Marketing: Joanna Gillette, B.A.
Senior Managing Editor: Edward Alderman, B.S., B.A.
Senior Medical Editor: Kimma Sheldon-Old, Ph.D., M.A.

For ordering information or questions, write or call:


Pharmacotherapy Self-Assessment Program
American College of Clinical Pharmacy
13000 W. 87th St. Parkway
Lenexa, KS 66215-4530
Telephone: (913) 492-3311
E-mail: accp@accp.com

PSAP 2022 Book 1 (Cardiology)


Library of Congress Control Number: 2021924037
ISBN-13s: 978-1-952291-37-1 (print); 978-1-952291-38-8 (eBook)

Copyright ©2022 by the American College of Clinical Pharmacy. All rights reserved. This book is protected by copyright. No part
of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, electronic or
mechanical, including photocopy, without prior written permission of the American College of Clinical Pharmacy.

Print versions are produced in the United States of America.

To cite PSAP properly:

Chapter authors. Chapter name. In: Sanoski CA, Witt DM, eds. Pharmacotherapy Self-Assessment Program, 2022 Book 1.
Cardiology. Lenexa, KS: American College of Clinical Pharmacy, 2022:page range.

PSAP™ is a registered trademark of the American College of Clinical Pharmacy.

Pharmacotherapy
Self-Assessment
Program
TABLE OF CONTENTS
Cardiology I�������������������������������������������������������������������������� 1 Triple Antithrombotic Therapy�������������������������������������������������������� 98
Cardiology I Panel ������������������������������������������������������������������������������ 3 Antithrombotic Therapy in Transcatheter Aortic Valve
Implantation ������������������������������������������������������������������������������������ 103
Antithrombotic Therapy in MitraClip ������������������������������������������� 105
Heart Failure with Reduced Ejection Conclusion �������������������������������������������������������������������������������������� 107
Fraction References �������������������������������������������������������������������������������������� 108
By Ralph J. Riello III, Pharm.D., BCPS Self-Assessment Questions�����������������������������������������������������������111
Introduction ���������������������������������������������������������������������������������������� 7
HF Updates According to the 2021 Expert Consensus
Cardiology III�������������������������������������������������������������������� 115
Decision Pathway ������������������������������������������������������������������������������ 9
Cardiology III Panel����������������������������������������������������������������������117
Optimizing GDMT ���������������������������������������������������������������������������� 15
Inotropic Therapy������������������������������������������������������������������������������ 17
Advanced Therapeutic Modalities ������������������������������������������������ 21 Management of Atrial Fibrillation
Heart Failure with Preserved Ejection Fraction �������������������������� 23 By Amy L. Lehnert, Pharm.D., BCPS, BCCP
Conclusion ���������������������������������������������������������������������������������������� 25
Introduction�������������������������������������������������������������������������������������� 121
References ���������������������������������������������������������������������������������������� 25
Definitions���������������������������������������������������������������������������������������� 122
Self-Assessment Questions������������������������������������������������������������ 29
Mechanisms������������������������������������������������������������������������������������ 122
Diagnostic Criteria�������������������������������������������������������������������������� 125
Drug-Induced Cardiovascular Disease Clinical Management of AF������������������������������������������������������������ 125
By Katherine Aymond, Pharm.D., BCPS, BCCP Contemporary Strategies for Stroke Prevention������������������������ 138
Contemporary Strategies for Rhythm Control���������������������������� 141
Introduction �������������������������������������������������������������������������������������� 33
AF Management in Special Populations�������������������������������������� 148
Chemotherapy and Immunotherapy-Induced Cardiotoxicity ������ 33
Conclusion �������������������������������������������������������������������������������������� 149
Other Agents of Cardiotoxicity ������������������������������������������������������ 42
References �������������������������������������������������������������������������������������� 149
Conclusion ���������������������������������������������������������������������������������������� 51
Self-Assessment Questions���������������������������������������������������������� 152
References ���������������������������������������������������������������������������������������� 51
Self-Assessment Questions������������������������������������������������������������ 53
Non-Statin Therapy for Dyslipidemia
By Nicholas W. Carris, Pharm.D., BCPS; and Kevin Cowart,
Cardiology II ���������������������������������������������������������������������� 57
Pharm.D., MPH, BCACP, CDCES
Cardiology II Panel���������������������������������������������������������������������������� 59
Introduction�������������������������������������������������������������������������������������� 157
Populations Likely to Benefit from Non-Statin Therapy������������ 158
Peripheral Arterial Disease
Non-Statin Agents for Dyslipidemia �������������������������������������������� 166
By Anastasia L. Armbruster, Pharm.D., FACC, BCPS, BCCP Emerging Non-Statin Therapies: Medications
Introduction �������������������������������������������������������������������������������������� 63 in the Pipeline���������������������������������������������������������������������������������� 172
Diagnosis ������������������������������������������������������������������������������������������ 65 Implementing Non-Statin Therapy ���������������������������������������������� 173
Pharmacotherapy in Patients with PAD ���������������������������������������� 66 Conclusion �������������������������������������������������������������������������������������� 174
Symptomatic Management ������������������������������������������������������������ 71 References �������������������������������������������������������������������������������������� 174
Critical Limb Ischemia �������������������������������������������������������������������� 72 Self-Assessment Questions���������������������������������������������������������� 177
Acute Limb Ischemia ���������������������������������������������������������������������� 73
Conclusion ���������������������������������������������������������������������������������������� 75
Cardiology IV ������������������������������������������������������������������ 181
References ���������������������������������������������������������������������������������������� 76
Cardiology IV Panel������������������������������������������������������������������������ 183
Self-Assessment Questions������������������������������������������������������������ 79

Interactive Case: Hyperlipidemia


Antithrombotic Therapy in Cardiac Management for Special Populations
Interventions
By Glenn Herrington, Pharm.D., AACC, BCCP, BCPS, CDCES;
By Stephanie Dwyer Kaluzna, Pharm.D., BCCP; and Daniel M. Riche, Pharm.D., FCCP, CLS, ASH-CHC
and Jaclynne Gowen, Pharm.D., BCCP Interactive Case: Hyperlipidemia Management
Percutaneous Coronary Intervention �������������������������������������������� 83 for Special Populations������������������������������������������������������������������ 187
Use of P2Y 12 Inhibitor in Percutaneous Coronary Hyperlink to activity������������������������������������������������������������������������ 188
Intervention �������������������������������������������������������������������������������������� 85 Self-Assessment Questions���������������������������������������������������������� 191

PSAP 2022 Book 1 • Cardiology iii Table of Contents


Interactive Case: Anticoagulation in Special Interactive Case: Cardiovascular Diseases
Populations in Pregnancy
By Allison E. Burnett, Pharm.D., CACP; and Kelly M. Rudd, By Lindsey Federle, Pharm.D., BCCP, BCPS
Pharm.D., FCCP, BCPS, CACP Interactive Case: Cardiovascular Diseases in Pregnancy ��������207
Interactive Case: Anticoagulation in Special Populations�������� 195 Hyperlink to activity������������������������������������������������������������������������208
Hyperlink to activity������������������������������������������������������������������������ 196 Self-Assessment Questions���������������������������������������������������������� 210
Self-Assessment Questions����������������������������������������������������������202

PSAP 2022 Book 1 • Cardiology iv Table of Contents


Cardiology I
Cardiology I Panel

Series Editors: Drug-Induced Cardiovascular Disease


Cynthia A. Sanoski, Pharm.D., FCCP, BCPS Author
Department Chair Katherine Aymond, Pharm.D., BCPS, BCCP
Associate Professor of Pharmacy Practice
Clinical Assistant Professor
Thomas Jefferson University
School of Clinical Sciences
Philadelphia, Pennsylvania
University of Louisiana Monroe
Daniel M. Witt, Pharm.D., FCCP, BCPS Clinical Pharmacy Specialist - ICU
Professor and Chair Department of Pharmacy
Department of Pharmacotherapy University Medical Center New Orleans
Assistant Dean of Clinical Affairs New Orleans, Louisiana
University of Utah College of Pharmacy
Salt Lake City, Utah Reviewers
Barbara S. Wiggins, Pharm.D., MBA, FCCP,
Faculty Panel Chair: FACC, FNLA, BCPS, BCCP, BCCCP, CLS
Craig J. Beavers, Pharm.D., FCCP, FACC, FAHA, BCCP, Clinical Pharmacy Specialist - Cardiology
BCPS-AQ Cardiology, CACP Department of Pharmacy
Director of Cardiovascular Services Medical University of South Carolina
Baptist Health Paducah Affiliate Professor
Cardiovascular Clinical Pharmacist Medical University of South Carolina School of Pharmacy
Department of Pharmacy Services Charleston, South Carolina
UK Healthcare Susan M. Smith, Pharm.D., BCPS
Assistant Adjunct Professor
Associate Professor of Pharmacy
Department of Pharmacy Practice and Science
School of Pharmacy
University of Kentucky College of Pharmacy
Wingate University
Paducah, Kentucky
Wingate, North Carolina

Heart Failure with Reduced Ejection


Fraction
Author
Ralph J. Riello III, Pharm.D., BCPS
Clinical Pharmacy Specialist, Clinical & Translational
Research
Department of Internal Medicine
Yale University School of Medicine
New Haven, Connecticut

Reviewers
Stormi E. Gale, Pharm.D., BCPS, BCCP
Assistant Professor
Department of Pharmacy Practice and Science
University of Maryland School of Pharmacy
Baltimore, Maryland
Nilam R. Naik, Pharm.D., BCPS, CACP
Clinical Pharmacy Specialist
VA Tennessee Valley Healthcare System
Nashville, Tennessee
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
Consultancies: Anastasia Armbruster (ACC/AHA Joint Committee Clinical Practice Guidelines); Jessica Bente (ACCP); Allison
Burnett: Consultancies (Anticoagulation Forum; National Certification Board for Anticoagulation Providers; Global Strategies);
Jessie L. Dunne (Pulmonary Hypertension Association); Nancy M. Nix (Pharmacosmos); Daniel M. Riche (Novo Nordisk; Merck;
AstraZeneca); Ralph J. Riello III (AstraZeneca; Johnson & Johnson); Sara Vazquez (Anticoagulation Forum);

Stock Ownership:

Royalties: Allison Burnett (Wolters-Kluwer); Daniel M. Riche (McGraw-Hill); Honoraria (Merck); Sara Vazquez (UpToDate)

Grants: Anastasia Armbruster (ACCP); Jessica Bente (New Jersey Society of Health-System Pharmacists); Gregory Castelli
(American Board of Family Medicine); Kevin Cowart (University of South Florida Taneja College of Pharmacy); Ralph J. Riello
III (AstraZeneca)

Honoraria: Anastasia Armbruster (AstraZeneca); John Bucheit (Association of Diabetes Care & Education Specialists); Nancy M.
Nix (Coherus; Pfizer; AstraZeneca); Ralph J. Riello III (Alexion; AstraZeneca; Janssen)

Other:

Nothing to disclose: Katherine Aymond, Eugene N. Bush, Nicholas W. Carris, Stephanie Dwyer Kaluzna, Lindsey Federle, Beth-
any A. Ford, Stormi E. Gale, Jaclynne R. Gowen, Glenn Herrington, Christine Ji, Amy L. Lehnert, Nilam R. Naik, Brenda Pahl, Joel
J. Peterson, A. Joshua Roberts, Kelly M. Rudd, Ashley Schenk, Susan M. Smith, Elisabeth M. Wang, Toby C. Trujillo, Barbara Wig-
gins, Leslie Wooten, Eman El Sayed Younis

ROLE OF BPS: The Board of Pharmacy Specialties (BPS) is an autonomous division of the American Pharmacists Association
(APhA). To maintain its strict, independent standards for certification, BPS does NOT endorse or provide review information,
preparatory courses, or study guides for Board Certification Examinations. The Board, through its specialty councils, is respon-
sible for specialty examination content, administration, scoring, and all other aspects of its certification programs. BPS is totally
separate and distinct from ACCP. PSAP has been approved by BPS for use in BCPS recertification. Information about the BPS
recertification process is available online.

Questions regarding BPS specialty recertification should be directed to:

Board of Pharmacy Specialties


2215 Constitution Avenue NW
Washington, DC 20037
(202) 429-7591
Continuing Pharmacy Education
and Recertification Instructions
Continuing Pharmacy Education (CPE) Credit: The American College of Clinical Pharmacy is accredited by the
Accreditation Council for Pharmacy Education (ACPE) as a provider of CPE.

PSAP Target Audience: The target audience for PSAP 2022 Book 1 (Cardiology) is pharmacotherapy specialists and advanced
level clinical pharmacists encountering diverse cardiovascular patient populations.

Available CPE credits: Purchasers who successfully complete all posttests for PSAP 2022 Book 1 (Cardiology) can earn 21.5 contact
hours of CPE credit. The universal activity numbers are as follows: Cardiology I – 0217-0000-22-002-H01-P, 5.0 contact hours;
Cardiology II – 0217-0000-22-003-H01-P, 6.0 contact hours; Cardiology III – 0217-0000-22-004-H01-P, 5.0 contact hours; and
Cardiology IV – 0217-0000-22-005-H01-P, 5.5 contact hours. You may complete one or all available modules for credit. Tests may
not be submitted more than one time.

TO EARN CPE CREDITS FROM THIS PSAP BOOK

Posttest access: Go to www.accp.com and sign in with your e-mail address and password. Technical support is available from 8 a.m.
to 5 p.m. (Central) weekdays by calling (913) 492-3311. PSAP products are listed under My Products on your My Account page.

BCPS test deadline: 11:59 p.m. (Central) on July 15, 2022.


ACPE test deadline: 11:59 p.m. (Central) on January 18, 2025.

BCPS Recertification Credit: To receive BCPS recertification CPE credit, a PSAP posttest must be submitted within
the 6-month period after the book’s release (see above). Only completed tests are eligible for credit; no partial
or incomplete tests will be processed. You may complete one or all available modules for credit. Tests may not be
submitted more than one time.

The passing point to earn BCPS recertification credit is based on an expert analysis of the assessment items in each posttest
module. Any posttest submitted before the BCPS test deadline that meets this passing point will earn BCPS recertification
credits. These credits will be assigned as of the date of test submission and reported within 48 hours to BPS. For statements of
recertification credit, visit www.bpsweb.org.

Questions regarding the number of hours required for BCPS recertification should be directed to BPS at (202) 429-7591 or
www.bpsweb.org. The ACCP Recertification Dashboard is a free online tool that can track recertification credits as they are
earned through ACCP and schedule new opportunities for credits from upcoming ACCP professional development programs.

ACPE CPE Credit: To receive ACPE CPE credit for a PSAP module, a posttest must be submitted within 3 years after the book’s
release (see above). Only completed tests are eligible for credit; no partial or incomplete tests will be processed. You may complete
one or all available modules for credit. Tests may not be submitted more than one time.

Any posttest submitted before the ACPE deadline that scores 50% or greater will be awarded the appropriate CPE. These
credits will be assigned as of the date of test submission and reported within 48 hours. For statements of CPE credit, visit
www.mycpemonitor.net.

Posttest answers: The explained answers—with rationale and supporting references—will be posted 2 weeks after the BCPS
test deadline and will be available to anyone who has either (1) submitted a posttest or (2) waived the right to receive credit from
a posttest (see below). Go to www.accp.com and sign in with your e-mail address and password. Click the PSAP book on your
My Account page and you will see a link to the explained answers.

Test Waivers: To access the explained answers without submitting a posttest, sign in to your My Account page, select the PSAP
book, and click on the waiver link for that module. By completing the waiver form for a module, you waive the opportunity to
receive CPE credit for that module. After you submit a waiver, you will see a link to the PDF file that contains the answers for the
module you waived. Answers will be available starting 2 weeks after the BCPS test deadline.
Heart Failure with Reduced Ejection
Fraction
By Ralph J. Riello III, Pharm.D., BCPS

Reviewed by: Stormi E. Gale, Pharm.D., BCPS, BCCP; and Nilam R. Naik, Pharm.D., BCPS, CACP

LEARNING OBJECTIVES

1. Distinguish phenotypic, structural, and functional classifications of heart failure to assess the stage of illness and delay
disease progression.
2. Justify the incorporation of recently approved pharmacologic therapies for heart failure with reduced ejection fraction
(HFrEF) into evidence-based therapies consistent with the American College of Cardiology’s 2021 Expert Consensus
Decision Pathway.
3. Develop a pharmacologic treatment plan for HFrEF that optimizes the use of traditional guideline-directed medical therapy.
4. Evaluate the role of inotropic agents and advanced therapeutic modalities available for patients with stage D heart failure.
5. Assess the potential benefit of pharmacologic therapies with recently expanded indications and promise for treatment
of heart failure with preserved ejection fraction.

INTRODUCTION
ABBREVIATIONS IN THIS CHAPTER
Epidemiology and Public Health Burden of Heart
ACC/AHA American College of Cardiology/
American Heart Association Failure
ACEI Angiotensin-converting enzyme Heart failure (HF) is a staggering public health burden in the United
inhibitor States, affecting individual patients, caregivers, clinicians, and health
ARB Angiotensin receptor blocker care systems across the country. Nearly 1 million new cases of HF are
CKD Chronic kidney disease diagnosed each year, amounting to a national prevalence of about 6
ECDP Expert Consensus Decision million Americans aged 20 years or older (Virani 2021). Although con-
Pathway temporary health policy efforts aim to improve care efficiency and
eGFR Estimated glomerular filtration rate delay the progression of disease, the aging population is projected
GDMT Guideline-directed medical therapy to drive a 46% increase in HF prevalence by 2030, which will affect
HF Heart failure more than 8 million adults, or 3.0% of the general population (Gerber
HFimpEF Heart failure with improved 2015). Heart failure is evenly proportioned between HF with reduced
ejection fraction
ejection fraction (HFrEF) and HF with preserved ejection fraction
HFmrEF Heart failure with mildly reduced
(HFpEF), with 53% of patients having impaired systolic function and
ejection fraction
the remaining 47% having preserved systolic function. However, gen-
HFpEF Heart failure with preserved ejec-
tion fraction der and racial disparities persist in HF consistent with cardiovascu-
HFrEF Heart failure with reduced ejection lar disease as a whole: Black men are more commonly hospitalized
fraction with HFrEF at 70% and white women at 59%; those two groups consti-
HFSA Heart Failure Society of America tute the highest proportion of HFpEF hospitalizations (Virani 2021).
HHF Hospitalization for heart failure Despite long-standing availability of lifesaving pharmacotherapies,
JHFS Japanese Heart Failure Society there remains a pervasive underutilization of evidence-based HFrEF
LVEF Left ventricular ejection fraction medications, whereas only limited disease-modifying treatment
MRA Mineralocorticoid receptor options are available for patients with HFpEF (Greene 2018). Con-
antagonist sequently, the 5-year mortality rate of overall HF rivals most of the
NT-proBNP N-terminal fragment B-type major malignancies—at 52.6%—whereas 1-year mortality reaches a
natriuretic peptide strikingly high 29.6% (Gerber 2015).

PSAP 2022 Book 1 • Cardiology 7 Heart Failure with Reduced Ejection Fraction
RAAS Renin-angiotensin-aldosterone system In addition to alarming rates of morbidity and mortality,
SGLT2 Sodium-glucose cotransporter 2 HF consistently ranks as the costliest condition in the United
T2DM Type 2 diabetes mellitus States, with expenditures totaling $30.7 billion—two-thirds
of which is attributable to direct medical costs (Heidenreich
Table of other common abbreviations. 2013). Because of the 30-day rehospitalization rate of 18.2%
among Medicare beneficiaries with HF, much of the cost bur-
den disproportionately affects acute-care facilities (Virani
2021). The tremendous strain HF exerts on health care insti-
tutions across the country has influenced payment reform to
BASELINE KNOWLEDGE STATEMENTS incentivize improved coordination of HF care delivery at the
system level. Though significant health policy advancements
Readers of this chapter are presumed to be familiar
have been made during the past decade, little or no improve-
with the following:
ment in hospital readmission rates or 30-day mortality has
• General understanding of the pathophysiologic yet been realized. It is therefore imperative that a standard-
derangements leading to heart failure
ized approach to the risk stratification, diagnosis, and staging
• Knowledge of the American College of Cardiology/ of HF progression be adopted so as to more readily identify
American Heart Association staging system and
the New York Heart Association functional classifi- patients appropriate for implementation of disease-modify-
cation of heart failure ing pharmacotherapy and advanced therapeutic modalities.
• Familiarity with traditional pharmacologic
therapies indicated for HFrEF, including angiotensin- Distinguishing Between the Different
converting enzyme inhibitors, angiotensin receptor Definitions and Classifications of Heart Failure
blockers, β-blockers, and mineralocorticoid recep- Previous definitions of HF were highly ambiguous and incon-
tor antagonists
sistent across varying platforms, with indiscriminate focus
Table of common laboratory reference values on hemodynamic parameters, pathophysiologic aspects,
and other clinical diagnostic features. Patients, clinicians,
and investigators facing an unclear picture of HF despite
ADDITIONAL READINGS
the growing epidemiologic burden of disease underscore
The following free resources have additional back- the importance of an updated, standardized characteriza-
ground information on this topic: tion of the illness. A recently proposed universal definition
• Maddox TM, Januzzi JL, Allen LA, et al. 2021 Update as set forth by the Heart Failure Society of America (HFSA),
to the 2017 ACC Expert Consensus Decision the Heart Failure Association of the European Society of Car-
Pathway for Optimization of Heart Failure diology (HFA-ESC), and the Japanese Heart Failure Society
Treatment: Answers to 10 Pivotal Issues About (JHFS) describes HF as clinical syndrome with a specific con-
Heart Failure With Reduced Ejection Fraction: stellation of symptoms and structural or functional cardiac
A Report of the American College of Cardiology
abnormalities known to be associated with HF. Those cardinal
Solution Set Oversight Committee. J Am Coll
Cardiol 2021;77:772-810. symptoms include dyspnea, fluid retention or edema, fatigue,
and intolerance of daily-life activities. The physical presen-
• Yancy CW, Jessup M, Bozkurt B, et al. 2017 ACC/ tation of HF must be further corroborated by the presence
AHA/HFSA focused update of the 2013 ACCF/AHA
guideline for the management of heart failure: a of elevated biomarkers or objective evidence of cardiogenic
report of the American College of Cardiology/ congestion. More specifically, elevated B-type-natriuretic-
American Heart Association Task Force on Clinical peptide levels of 35 or more or 100 or more pg/mL or N-terminal
Practice Guidelines and the Heart Failure Society fragment proBNP (NT-proBNP) of 125 or more or 300 or more
of America. Circulation 2017;136:e137-61.
pg/mL for the ambulatory or hospital setting, respectively,
• McDonagh TA, Metra M, Adamo M, et al. 2021 ESC must be present alongside confirmatory signs of HF captured
guidelines for the diagnosis and treatment of acute
by means of diagnostic modalities such as chest radiogra-
and chronic heart failure: developed by the Task
phy, echocardiography, or right heart catheterization (Boz-
Force for the Diagnosis and Treatment of Acute
and Chronic Heart Failure of the European Society kurt 2021). Notably, natriuretic peptide thresholds endorsed
of Cardiology (ESC) with the special contribution of by the HFSA/HFA-ESC/JHFS for HF diagnosis—although
the Heart Failure Association (HFA) of the ESC. consistent with clinical practice guidelines—may have lower
Eur Heart J 2021;42:3599-726. specificities in patients with advanced age, atrial fibrillation,
• Neu R, Leonard MA, Dehoorne ML, et al. Impact of or chronic kidney disease (CKD).
pharmacist involvement in heart failure transition Beyond an updated definition of disease, new categories of
of care. Ann Pharmacother 2020;54:239-46. HF according to left ventricle ejection fraction (LVEF) were also
recently developed (Table 1). Both HFrEF and HFpEF remain

PSAP 2022 Book 1 • Cardiology 8 Heart Failure with Reduced Ejection Fraction
disease description and new LVEF categories. The original
Table 1. Universal Classification of HF According American College of Cardiology/American Heart Associa-
to LVEF tion (ACC/AHA) staging system that described HF was based
entirely on symptoms and the unidirectional absence or pres-
HFSA/HFA-ESC/
JHFS classification LVEF ence of structural heart disease. Although it may be widely
known among clinicians, the ACC/AHA platform does not
HFrEF ≤40%
incorporate the evolving role of biomarkers in disease pro-
HFmrEF 41% – 49% gression, nor does it exhibit strong association with prog-
HFpEF ≥50% nosis or quality of life. The recent HFSA/HFA-ESC/JHFS
classification of HF revises the previous ACC/AHA staging
HFimpEF Baseline ≤40%, a ≥10-point
system to address the prior approach’s gaps and limitations
increase from baseline and a
and to enhance patient and public understanding and adop-
second measurement of >40%
tion (Bozkurt 2021). The revised HFSA/HFA-ESC/JHFS stag-
ing platform recognizes HF as a continuum of illness with
HFimpEF = heart failure with improved ejection fraction;
HFmrEF = heart failure with mildly reduced ejection frac- corresponding clinical trajectories as patients move from
tion; HFpEF = heart failure with preserved ejection fraction; at risk of HF to pre-HF or de novo HF to more-advanced dis-
HFrEF = heart failure with reduced ejection fraction; HFSA/ ease. The New York Heart Association’s (NYHA’s) functional
HFA-ESC/JHFS = Heart Failure Society of America/Heart
classification of HF offers a complementary understand-
Failure Association of the European Society of Cardiology/
Japanese Heart Failure Society; LVEF = left ventricular ing of a patient’s ACC/AHA or HFSA/HFA-ESC/JHFS stage
ejection fraction. by describing symptomatic severity as it affects limitations
Information from: Bozkurt B, Coats AJS, Tsutsui H, et al. on the activities of daily life (Table 2). Unlike the ACC/AHA
Universal definition and classification of heart failure: a staging system, the NYHA scale is a bidirectional functional
report of the Heart Failure Society of America, Heart
assessment and exhibits strong correlation with mortality as
Failure Association of the European Society of Cardiology,
Japanese Heart Failure Society and Writing Committee of well as health-related quality of life. Pharmacists should be
the Universal Definition of Heart Failure. J Card Fail 2021; become familiar with both HFSA/HFA-ESC/JHFS staging and
27:P387-413. NYHA classifications so as to become able to identify appro-
priate pharmacotherapy commensurate with illness sever-
ity to delay disease progression, reduce mortality risk, and
defined as LVEF 40% or less and 50% or more, respectively. improve quality of life.
However, the dichotomy of LVEF above or below the traditional
40% threshold has been expanded to include HF with mildly
HF UPDATES ACCORDING TO THE
reduced EF (HFmrEF), which distinguishes transitional patients
2021 EXPERT CONSENSUS DECISION
with LVEFs of 41% – 49% and which represents an underinves-
PATHWAY
tigated subgroup that comprises up to 20% of patients with HF
(Bozkurt 2021). Previously referred to as having midrange HF, Biomarker Considerations
patients with HFmrEF overlap the characteristics of both HFrEF In addition to providing practical guidance to integrate
and HFpEF—but they may be more likely to benefit from neuro- recently approved pharmacotherapeutic classes into the
hormonal antagonism than would patients with preserved EF. care of patients with HFrEF, recent recommendations also
Although HFmrEF may indicate early signs of deteriorating sys- highlight the importance of the routine incorporation of bio-
tolic dysfunction, this classification can also reflect recovering markers into clinical practice as well as management strat-
EF—typically in the setting of adherence to disease-modifying egies for common comorbid conditions (Maddox 2021).
therapy. Therefore, periodic echocardiography assessment is The natriuretic peptide system is a central counterregula-
recommended as a way of monitoring trends in LVEF overtime tory process directly compensatory to HF pathophysiology.
and can meaningfully inform response to therapy. To more spe- In response to ventricular wall stretch caused by high intra-
cifically define the phenomenon, a new category was created cardiac filling pressures, atrial natriuretic peptide and BNP
to characterize patients with HF who also have LVEFs from get synthesized and then released from cardiac myocytes.
which they have indeed recovered. Heart failure with improved These neurohormones promote diuresis, natriuresis, and
EF (HFimpEF) represents such patients with a positive trajec- vasodilation while also inhibiting both the sympathetic ner-
tory phenotype. Patients with HFimpEF have baseline LVEFs of vous system and the renin-angiotensin-aldosterone system
40% or less, but they experience a 10-or-more-point increase (RAAS). B-type natriuretic peptide and its inactive proteolytic
in systolic function, with a subsequent measurement of more fragment, NT-proBNP, are the two most well-characterized
than 40% (Bozkurt 2021). biomarkers in HF. Specifically, clinical practice guidelines
Proposed revisions to stages in the development and confer a Class I recommendation to measure serum BNP or
progression of HF also extend further than only an updated NT-proBNP concentrations in order to establish or exclude a

PSAP 2022 Book 1 • Cardiology 9 Heart Failure with Reduced Ejection Fraction
Table 2. Comparison of Structural and Functional Classifications of HF Development and Progression

HFSA/HFA-ESC/JHFS Universal Definition


ACC/AHA HF Staging System and Classification of HF NYHA Functional Classification
Stage Description Stage Description Class Description

A Patients at high risk At risk Patients at risk of HF but without No associated N/A
of developing HF but current or prior symptoms or functional
without structural heart signs of HF and without structural, class
disease (e.g., HTN, biomarker, or genetic markers of heart
DM, CAD, metabolic disease (e.g., HTN, CVD, DM, obesity,
syndrome) known exposure to cardiotoxins,
cardiomyopathy history)

B Patients with structural Pre-HF Patients without current or prior No associated N/A
heart disease but no symptoms or signs of heart failure functional
signs or symptoms of but having evidence of one of the class
HF (prior MI, low EF, following: structural heart disease
no symptoms) (LVH, valvular heart disease, chamber
enlargement, etc.), abnormal cardiac
function (reduced LV or RV systolic
function, increased filling pressures,
etc.), elevated natriuretic peptide
levels (or elevated cardiac troponin
levels after cardiotoxin exposure)

C Patients with structural HF Patients with current or prior I No limitations of


heart disease and symptoms and/or signs of HF caused physical activity
current or previous by structural and/or functional cardiac
II Slight limitation of
symptoms (low EF, abnormalities
physical activity
HF signs/symptoms)
III Marked limitation
of physical activity

D Patients with symptoms Advanced Severe symptoms and/or signs of HF IV Unable to carry
despite maximal medical HF at rest, recurrent hospitalizations on any physical
therapy (end-stage HF) despite GDMT, refractory to or activity without
intolerant of GDMT requiring discomfort
advanced therapies

CAD = coronary artery disease; CVD = cardiovascular disease; DM = diabetes mellitus; EF = ejection fraction; GDMT = guideline-
directed medical therapy; HF = heart failure; HTN = hypertension; LV = left ventricle; LVH = left ventricular hypertrophy; MI = myocar-
dial infarction; N/A = not applicable; NYHA = New York Heart Association; RV = right ventricle.
Information from: Bozkurt B, Coats AJS, Tsutsui H, et al. Universal definition and classification of heart failure: a report of the Heart
Failure Society of America, Heart Failure Association of the European Society of Cardiology, Japanese Heart Failure Society and
Writing Committee of the Universal Definition of Heart Failure. J Card Fail 2021;27:P387-413.

clinical diagnosis of HF, evaluate illness severity, and eluci- applied to assess responsiveness to guideline-directed med-
date overall prognosis (Yancy 2017). Elevated natriuretic pep- ical therapy (GDMT). This is because BNP and NT-proBNP
tide concentrations in an ambulatory patient with HFrEF, for concentrations typically get decreased by evidence-based
example, may be suggestive of an imminent risk of decom- pharmacotherapy proportional to their magnitude of clinical
pensation requiring intravenous diuretics and further escala- benefit. Conversely, patients with HFrEF whose natriuretic
tion of care—particularly if levels are acutely increased from peptide levels fail to improve despite adherence to GDMT
baseline. may be considered nonresponders, implying poor prognosis
Not only do natriuretic peptide levels play a prominent role and advanced illness. In the Guiding Evidence Based Therapy
in HF diagnosis and staging, but they also inform prognosis, Using Biomarker Intensified Treatment in HF trial, patients
determine risk stratification, and have more recently been with HFrEF who achieved goal NT-proBNP levels of less than

PSAP 2022 Book 1 • Cardiology 10 Heart Failure with Reduced Ejection Fraction
1,000 pg/mL were associated with significant reverse ventric- patients with HFrEFs. The condition is independently asso-
ular remodeling, improved LVEF, and fewer adverse events ciated with poor prognosis, reduced quality of life, and dimin-
after 1 year —independent of management strategy (Daubert ished exercise capacity irrespective of concomitant anemia
2019). Moreover, the Prospective Study of Biomarkers, (von Haehling 2019). Impaired oxygen delivery to tissue in
Symptom Improvement and Ventricular Remodeling During patients with anemia precipitates neurohormonal and hemo-
Entresto Therapy for Heart Failure Study demonstrated that dynamic derangements that may overlap and exacerbate
both the rapidity and robustness of NT-proBNP reductions symptoms of HF like fatigue and dyspnea. Although screen-
after angiotensin- receptor–neprilysin-inhibitor (ARNI) initi- ing for reversible causes of anemia is essential in a routine
ation were associated with corresponding improvements in baseline evaluation for HF, the etiology is often complex and
reverse remodeling as well as the odds of HF hospitalization multifactorial. Functional iron deficiency caused by inflam-
or death (Januzzi 2020). Consequently, routine monitoring mation in the setting of chronic illness is not well understood,
of natriuretic peptide concentrations serves as a useful risk but it is important to know that lower ferritin thresholds for
assessment and treatment responsiveness tool. However, diagnosis apply to individuals without chronic conditions.
specific natriuretic-peptide-level targets as treatment goals Decreased dietary iron intake and reduced ferrous absorp-
are not currently endorsed by guidelines (Maddox 2021). tion in the edematous gut wall are likely contributory, but
When using biomarkers to aid in clinical decision-making more-complex mechanisms related to iron sequestration
around intensification of GDMT, careful consideration must have also been identified. It is important to correct iron defi-
be given to assay interpretation for patients prescribed sacu- ciency in patients with HF, and therefore, pharmacists must
bitril/valsartan. Mechanistically—because of its inhibitory carefully consider the iron formulation, route of administra-
effect on neprilysin, a neutral endopeptidase responsible tion, and dosing regimen.
for BNP degradation—the sacubitril component of ARNI may Enteral iron preparations are poorly absorbed, often cause
cause concentrations of BNP to moderately increase and many unpleasant GI side effects, and require up to 6 months
thereby further delay a return to baseline levels. And because to replenish iron stores. The IRON-5 and IRONOUT-HF stud-
NT-proBNP is not a substrate for neprilysin, however, it may ies investigated the impact of ferrous sulfate and iron poly-
be preferable to BNP as a monitoring parameter in the con- saccharide, respectively, but failed to demonstrate any
text of ARNI treatment (Maddox 2021). Pharmacists engaged functional impact on peak oxygen consumption in patients
in biomarker monitoring in the acute- or ambulatory-care with iron-deficient HFrEF (von Haehling 2019). Intravenous
context should inform HF-treatment providers of this unique iron avoids many of the drawbacks associated with oral sup-
interaction and then guide interpretation as needed. plementation and appears to hold more promising clinical
benefit. Short-term exposure with parenteral ferric carboxy-
Comorbidity Management in HF maltose in the FAIR-HF and CONFIRM-HF trials improved
Although HF is the leading individual cause of hospitalization NYHA class performance and 6-minute walk-test perfor-
in the United States, the presence of either cardiovascular mance. Ferric carboxymaltose was associated with a lower
or noncardiac comorbidities significantly increases the risk HF hospitalization risk in CONFIRM-HF, but the study was
of further complications (Virani 2021). Multiple Class I and underpowered for assessment of clinical end points. Still,
III antiarrhythmics used for control of atrial and ventricular the 2017 ACC/AHA/HFSA guideline preferentially endorsed
arrhythmias, for example, are contraindicated in HF because intravenous iron replacement in NYHA class II to IV HF with
of their negative inotropic or proarrhythmic effects. In addi- concomitant deficiency (Yancy 2017). Subsequently, the Ran-
tion, widely prescribed oral hypoglycemics for type 2 diabe- domised, Double-Blind Placebo Controlled Trial Comparing
tes mellitus (T2DM)—such as thiazolidinediones often cause the Effect of Intravenous Ferric Carboxymaltose on Hospital-
edema, which can precipitate HF symptoms. Even OTC med- isations and Mortality in Iron Deficient Patients Admitted for
ications such as NSAIDs or nasal decongestants like phenyl- Acute Heart Failure (AFFIRM-AHF) study demonstrated a 26%
ephrine and pseudoephedrine can worsen HF (Pagell 2016). relative risk reduction in total HF hospitalizations with ferric
Guideline-directed medical therapy for HFrEF now also neces- carboxymaltose compared with placebo (217 [48.9%] vs. 294
sitates the use of four concomitant medication classes—all [53.5%]; HR 0.74; 95% CI, 0.58–0.94, p=0.013) when initiated
of which lower blood pressure. Therefore, prudent prescrib- before discharge in clinically stabilized patients with iron-
ing for patients with HF and comorbid conditions must be deficient acute HF with LVEFs of less than 50% after 1 year of
exercised so as to prevent polypharmacy and avoid medica- follow-up (Ponikowski 2020). It is notable that ferric carboxy-
tion-related adverse events. maltose was dosed at 500–2000 mg based on body weight
and hemoglobin; patients who were persistently deficient
Iron Deficiency received additional doses at weeks 6, 12, and 24 if needed.
Iron deficiency—characterized by a ferritin of less than 100 The ongoing FAIR-HF2, HEART-FID, and IRONMAN trials are
ng/mL or 100–300 ng/mL with transferrin saturation of less expected to further inform the role of parenteral iron repletion
than 20%—is estimated to affect approximately half of all

PSAP 2022 Book 1 • Cardiology 11 Heart Failure with Reduced Ejection Fraction
on clinical end points for patients with iron-deficient HF (clin- be warranted (Riello 2021). Finerenone, a novel nonsteroidal
icaltrials.gov). MRA, has also demonstrated cardiorenal benefits, with a low
incidence of hyperkalemia-related treatment discontinuation
Chronic Kidney Disease (1.2%–2.3%) in diabetic kidney disease studies FIDELIO-DKD
The heart and kidney maintain saltwater homeostasis and and FIGARO-DKD (Pitt 2021; Bakris 2020). Recently, finere-
regulate blood pressure by way of interdependent neurohor- none received regulatory approval to reduce the risk of sus-
monal mechanisms that are critical to the function of either tained eGFR decline, end-stage renal disease, cardiovascular
organ alone. Cardiorenal disease is, unsurprisingly, a preva- death, nonfatal myocardial infarction, and HF hospitalization
lent manifestation of comorbid illness among patients with in patients with comorbid CKD. The potential benefit of finere-
HF, because approximately half also suffer from CKD and vice none is being investigated in HFpEF as well by way of active
versa (Virani 2021). Not only does CKD worsen an already poor enrollment of patients in the ongoing FINEARTS-HF trial (clin-
prognosis of HF, but also mortality increases proportionally icaltrials.gov).
to the degree of coexistent renal insufficiency. It is import- It is important that SGLT2 inhibitors be considered con-
ant to note that randomized control trials establishing lifesav- traindicated in severe renal impairment per the prescribing
ing pharmacotherapy for HFrEF have consistently excluded label—but for a lack of A1C-lowering efficacy in diabetes man-
patients with severe renal dysfunction. Furthermore, this agement not explicitly because of safety concerns or rela-
complex but common cohort is less likely to be prescribed tionship to nondiabetic indications. The SGLT2 inhibitors
GDMT compared with patients without kidney disease (Hein have been safely studied in patients with eGFRs as low as 20
2019). Many of the guideline-recommended medications for mL/minute/1.73 m2 for HF or CKD and have received corre-
HF may affect kidney function, often necessitating renal sponding label updates. For example, dapagliflozin recently
dose adjustments and close monitoring. For example, RAAS received expanded regulatory approval to reduce the risk of
inhibitors can potentiate the risk of hyperkalemia or precip- kidney function decline, kidney failure, cardiovascular death,
itate acute kidney injury. Consequently, initial ARNI dosing and hospitalization for HF on the basis of the DAPA-CKD
should be reduced to 24/25 mg twice daily if estimated glo- trial (Heerspink 2020). In this study of patients with eGFRs
merular filtration rate (eGFR) is less than 30 mL/minute/1.73 of 25–75 mL/minute/1.73 m2 and urinary albumin-to-creati-
m2, whereas mineralocorticoid receptor antagonists (MRAs) nine ratios of 200–5000, dapagliflozin improved cardiorenal
are not recommended for use unless eGFR is more than outcomes, including all-cause mortality independent of dia-
30 mL/minute/1.73 m2 and serum potassium is less than betes status after a median 2.4 years of follow-up compared
5 mEq/L (Yancy 2017). with placebo. The ongoing EMPA-Kidney trial is anticipated
Hyperkalemia is a well-established adverse medication to corroborate those beneficial effects with empagliflozin in
reaction to RAAS inhibition that worsens with kidney disease patients with broader ranges of renal impairment inclusive of
and acts as a common barrier to the initiation or uptitration eGFR 20–90 mL/minute/1.73 m2 (clinicaltrials.gov).
of GDMT (Maddox 2021). Beyond diet modifications to reduce
potassium intake, novel potassium-binding resins such as Diabetes
patiromer sorbitex calcium and sodium zirconium cyclosili- Among the most commonly encountered comorbidities for
cate are now indicated for management of acute and chronic patients with HF, T2DM requires a strategic multidisciplinary
hyperkalemia. Adjunctive use of patiromer is associated with approach to optimal management of both conditions simulta-
improved MRA utilization in patients with chronic HF and histo- neously. Uncontrolled hyperglycemia typically leads to isch-
ries of hyperkalemia, but the impact on other RAAS inhibitors emic HF through atherosclerotic or hypertensive mechanisms
is not yet known (Pitt 2011). The ongoing DIAMOND, LIFT, and but can also precipitate a diabetic cardiomyopathy that often
OPERA-HF trials should serve to (1) further define the roles of manifests as diastolic dysfunction and, eventually, HFpEF
newer-generation potassium-binding resins to more broadly (Jia 2018). Consequently, the presence of T2DM increases the
prevent hyperkalemia while enabling GDMT optimization and risk of developing incident HF more than twofold among the
(2) clarify any potential benefit in HF outcomes (clinicaltrials. general population; also, poor glycemic control significantly
gov). Consideration of those agents in treating comorbid HF increases HF hospitalization risk and decreases overall sur-
and CKD should include attention to binding interactions vival among patients with established disease (Dunlay 2019).
with other concurrent medications, GI discomforts, magne- Despite the substantial risk of cardiovascular compli-
sium derangements with patiromer, and edema caused by cations in patients with T2DM, many commonly prescribed
increased sodium load with sodium zirconium cyclosilicate glucose-lowering agents have failed to consistently demon-
(Hein 2019). Until the role of novel potassium binders in HF strate improvements in macrovascular outcomes. And several
is better understood, individualized selection of specific antihyperglycemics have even demonstrated safety concerns
GDMT agents that may confer lower comparable levels of related to HF. Thiazolidinediones as a class are known to carry
hyperkalemia or acute renal insufficiency risk such as ARNI an FDA boxed warning with regard to causing or exacerbat-
or a sodium-glucose cotransporter 2 (SGLT2) inhibitor may ing HF, and dipeptidyl peptidase 4 (DPP4) inhibitors alogliptin

PSAP 2022 Book 1 • Cardiology 12 Heart Failure with Reduced Ejection Fraction
and saxagliptin received regulatory precautions for increased with HFrEF and T2DM. And because GLP-1 agonists exhibit
HF hospitalization risk. Other conventional oral hypoglyce- positive chronotropic effects that consistently increase
mics like sulfonylureas and glinides have (1) only limited pro- heart rate by 5–10 beats/minute, precaution should be taken
spective evidence to support their safety in the treatment of when used in patients with diabetes and HFrEF—particu-
HF and (2) mixed findings in observational studies (Dunlay larly patients who may not be optimally prescribed β-blocker
2019). In consideration of the more-consistent cardiorenal therapy.
benefits demonstrated by SGLT2 inhibitors and glucagonlike The first antihyperglycemic-medication class to demon-
peptide 1 (GLP-1) receptor agonists across multiple robust strate cardiorenal benefits among patients with T2DM at
cardiovascular outcome trials, the most-recent American Dia- high cardiovascular risk consisted of the SGLT2 inhibitors.
betes Association guidelines recommend preferential use of The EMPA-REG OUTCOME trial randomized 7020 patients
these agents for patients who have T2DM with established with T2DM and established ASCVD to empagliflozin or pla-
atherosclerotic cardiovascular disease (ASCVD) or multiple cebo, evaluating three-point major adverse cardiovascular
cardiac risk factors (ADA 2020). Additional preference should events as the primary composite end point. After a median
be given to SGLT2 inhibitors specifically proven to reduce the follow-up period of 3.1 years, empagliflozin lowered the risk
risk of worsening HF and cardiovascular death in patients of the primary outcome by a striking 38%; HF hospitalizations
with diabetic HF—particularly those with HFrEF (Das 2020). by 35%; and all-cause mortality by 32% (Zinman 2015). The
Several GLP-1 receptor agonists have demonstrated subsequent, CANVAS clinical trial program integrated results
reductions in major adverse cardiovascular events among of canagliflozin across two placebo-controlled studies inclu-
patients with T2DM and established ASCVD or multiple sive of 10,142 patients, with about 65% having prior ASCVD
high-risk features. In the 9340-patient LEADER trial, liraglu- and with the remainder at only high cardiovascular risk (Neal
tide significantly reduced the primary composite outcome 2017). The larger proportion of patients under primary pre-
of cardiovascular death, nonfatal myocardial infarction, and vention likely contributed to the achievement of a significant
stroke compared with placebo after 3.8 years of median reduction in the same three-point composite end point of
follow-up (Marso 2016a). Both of the smaller, SUSTAIN-6 and empagliflozin but not the individual component of cardiovas-
PIONEER 6 trials comparing injectable and oral semaglutide cular death alone. Further, the CANVAS trial was complicated
with placebo, respectively, were underpowered to determine by a signal for increased risk of lower-limb amputations—par-
superiority; however, each formulation was associated with ticularly of the toe or metatarsal in patients with advanced dia-
similar reductions in the primary three-point composite end betes who were not otherwise observed in other canagliflozin
point (Husain 2019; Marso 2016b). Neither exenatide nor lix- studies to date (Perkovic 2019). The largest SGLT2-inhibitor
isenatide in the EXSCEL and ELIXA trials, respectively, demon- cardiovascular safety study—DECLARE-TIMI 58—enrolled
strated statistically significant reductions in cardiovascular the smallest proportion of patients with T2DM, established
outcomes compared with placebo. Weekly injections of dula- ASCVD at 40.6%, and, consequently, did not demonstrate
glutide compared with placebo in 9901 patients with T2DM improvement in the same three-point composite outcome
predominantly at high cardiovascular risk improved the risk (Wiviott 2018). However, given the consistent reduction in
of cardiovascular death, nonfatal myocardial infarction, and HHF demonstrated by prior SGLT2-inhibitor trials, the study
stroke in the REWIND trial (Gerstein 2019). Notably, REWIND oversight committee amended the protocol to incorporate an
was the only GLP-1 receptor agonist cardiovascular safety additional primary end point of cardiovascular death or hos-
study thus far to prespecify urgent HF visits as a secondary pitalization for HF. Compared with placebo, dapagliflozin sig-
outcome—though no differences versus placebo were found. nificantly reduced the right of this composite end point (4.9%
The benefit of most GLP-1 agonists appears to be driven by a vs. 5.8%; HR 0.83; 95% CI, 0.73–0.95; p=0.005)—predomi-
reduction in cardiovascular death; a discernible impact on HF nantly because of a reduction in HF hospitalizations. Despite
events has yet to be observed for GLP-1 agonists in patients an enrollment of 8246 patients with diabetes and established
who have T2DM and various cardiovascular-risk profiles. The ASCVD, ertugliflozin did not improve the risk of major adverse
limited evidence for GLP-1 receptor agonists in patients with cardiovascular events compared with placebo in the VER-
HF but without diabetes has been largely neutral, with one TIS-CV trial (Cannon 2020). In alignment with prior SGLT2-in-
notable exception. The FIGHT trial—which evaluated liraglu- hibitor trials, only HF hospitalizations were significantly
tide compared with placebo in 300 recently decompensated reduced with ertugliflozin. A dual SGLT1 and SGLT2 receptor
patients with HF irrespective of T2DM diagnosis—did not antagonist—sotagliflozin—is currently being considered for
determine any benefit for a variety HF-related outcomes and regulatory approval. In SCORED—a recent, 10,584-patient car-
functional status; however, a nonsignificant trend toward a diovascular outcomes study of patients with comorbid T2DM
numerically increased risk of death or HF hospitalization was and CKD and with or without albuminuria—sotagliflozin sig-
identified (HR 1.30; 95% CI, 0.92–1.83, p=0.14) throughout the nificantly reduced its amended coprimary composite end
6-month study duration (Margulies 2016). And that risk was point of cardiovascular death, HF hospitalizations, and urgent
higher (HR 1.54; 95% CI, 0.97–2.46, p=0.07) among patients HF visits (HR 0.74; 95% CI, 0.63–0.88; p<0.001) (Bhatt 2021a).

PSAP 2022 Book 1 • Cardiology 13 Heart Failure with Reduced Ejection Fraction
In the subsequent, SOLOWIST-WHF trial, which enrolled 1222 neither cardiovascular death (10% vs. 10.8%; HR, 0.92; 95% CI,
patients with T2DM and recent hospitalizations for worsen- 0.75-1.12) nor overall mortality (10.1% vs. 10.7%; HR, 0.92; 95%
ing HF, sotagliflozin also produced the same primary com- CI, 0.62-1.19) achieved statistically significant differences.
posite outcome (51.0% vs. 76.3%; HR 0.67; 95% CI, 0.52–0.85; An ensuing meta-analysis of both pivotal trials, however, did
p<0.001) compared with placebo (Bhatt 2021). Early termina- indeed suggest the benefits of dapagliflozin and empagli-
tions of the trials—caused by loss of sponsor funding and the flozin consistently improved a host of cardiorenal outcomes
advent of the COVID-19 pandemic—may have encumbered the in HFrEF patients independent of T2DM (Zannad 2020).
ability to detect differences in individual components of the The immense benefit of SGLT2 inhibitors in patients with
primary outcome. However, a pooled analysis of both studies or without diabetes is anticipated to rapidly increase overall
suggests the benefits of sotagliflozin may apply to patients prescribing rates, warranting practical considerations for rou-
with diabetes and HF irrespective of LVEF, including HFmrEF tine incorporation into the HF armamentarium. It is import-
and HFpEF (Bhatt 2021b). ant to note that the doses of dapagliflozin and empagliflozin
studied for HFrEF were the same, 10-mg once-daily regimen.
Dosing of empagliflozin should be increased to 25 mg only if
New Evidence-Based Medications for HFrEF
additional glucose lowering is desired—for example, in uncon-
SGLT2 Inhibitors trolled comorbid T2DM. Hemoglobin A1C reduction is unlikely
The use of SGLT2 inhibitors for cardiovascular-risk reduction to occur in advanced kidney disease, but cardiorenal bene-
in patients with T2DM has been part of consensus practice fits appear to be maintained if eGFR is 25 or more or 20 or
for the past few years, but their emerging role as the fourth more mL/minute/1.73 m2 with dapagliflozin and empagli-
pillar of HFrEF GDMT has been demonstrated only recently flozin, respectively. Nondiabetic patients—and even dia-
(Maddox 2021). By promoting glucosuria through a block- betic patients not concomitantly prescribed a sulfonylurea
ade of sodium and glucose reabsorption in the proximal or insulin therapy—rarely experience hypoglycemic events
tubule of the nephron, SGLT2 inhibitors lower glucose. Their from an SGLT2 inhibitor alone. To avoid ketosis, however,
mechanism for cardiorenal protection is not well understood (1) SGLT2 inhibitors should be held 3 days before major sur-
but appears to be multifactorial and related principally to gery in patients with T2DM and (2) confirmed modifiable risk
(1) hemoconcentration, (2) osmotic diuresis and natriuresis, factors for ketoacidosis should be resolved before reinitiat-
(3) arterial pressure and stiffness reduction, and (4) efficient ing. In addition, fluctuating clinical features such as renal
myocardial ketone metabolism—all of them independent of function, volume status, and nutritional intake should be eval-
hyperglycemia (Zelniker 2020). uated before routine inpatient continuation of chronic therapy
The first landmark randomized, controlled trial to establish or new initiation in hospitalized patients across therapeutic
the benefit of SGLT2 inhibitors beyond T2DM investigated the indications. Dose reductions of concurrent loop diuretic ther-
role of dapagliflozin in patients with HFrEF. In the DAPA-HF apy may be warranted over time for patients prescribed an
study, dapagliflozin reduced the risk of the primary composite SGLT2 inhibitor—especially if blood pressure is tenuous or
end point of worsening HF or cardiovascular death compared indicative of intravascular volume contraction. Although gen-
with placebo in 4744 patients with HFrEF and with or without ital mycotic infections are the most common adverse reac-
diabetes (16.3% vs. 21.2%; HR 0.74; 95% CI, 0.65–0.85) after tions to SGLT2 inhibitors—occurring predominantly in women
18.2 months of median follow-up (McMurray 2019). Unlike the and uncircumcised males—proper personal hygiene may mit-
previous, DECLARE-TIMI 58 study, dapagliflozin also signifi- igate risk. If treatment is required, topical or systemic anti-
cantly reduced the risk of key secondary end points, includ- fungal therapy typically alleviates discomfort. Urinary tract
ing cardiovascular death (9.5% vs. 11.5%; HR 0.82; 95% CI, infections occur less often but may be more likely to occur in
0.69–0.98) and all-cause mortality (11.6% vs. 13.9%; HR 0.83; patients with histories of pyelonephritis, kidney stones, uret-
95% CI, 0.71–0.97). Based on the landmark findings from the eral stenting, or indwelling urinary catheters.
DAPA-HF study, dapagliflozin received expanded regulatory
approval to reduce the risk of death or HF hospitalization sGC Modulators
among patients with HFrEF irrespective of the presence or The nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic
absence of T2DM. The EMPEROR-Reduced trial corroborated guanosine monophosphate (cGMP) pathway regulates vaso-
the benefits of SGLT2 inhibitors in patients with HFrEF and dilation and myocardial demand via smooth muscle relax-
diabetes and patients with HFrEF but without diabetes alike. ation. In HF, inadequate cardiac output and decreased NO
In the study, empagliflozin significantly reduced the primary bioavailability result in compensatory vasoconstriction and
composite outcome of HF hospitalization or cardiovascular reduced cGMP generation. Organic nitrate supplementation
death (19.4% vs. 24.7%; HR 0.75; 95% CI, 0.65–0.86) among activates NO-sGC-cGMP but is limited by tachyphylaxis and
3730 patients with HFrEF compared with placebo after a may contribute to oxidative stress. Novel oral sGC activators
median 16 months (Packer 2020). This benefit appeared to stimulate cGMP synthesis, enhance sGC sensitivity to NO,
be largely influenced by the effect on HF hospitalizations, as and promote vasodilation independent of NO or heme.

PSAP 2022 Book 1 • Cardiology 14 Heart Failure with Reduced Ejection Fraction
Vericiguat is the first sGC activator approved to reduce the recent decompensation despite optimal therapy for whom an
risk of cardiovascular death, hospitalization for heart failure additional branded medication does not cause undue finan-
(HHF), and outpatient intravenous diuretics in symptomatic cial burden. Further benefit may also be derived in patients
patients with chronic HF and LVEFs of less than 45%. Despite with low circulating biomarker levels, including cGMP, NO, and
recent regulatory approval, vericiguat initially disappointed BNP, although this precision-medicine approach to vericiguat
in the phase II, dose-finding Soluble Guanylate Cyclase therapy requires further elucidation to support routine con-
Stimulation in Heart Failure with Reduced Ejection Fraction sideration. Because of the exclusion of patients prescribed
(SOCRATES-REDUCED) trial (Gheorghiade 2015). The SOCRA- long-acting nitrates and NO donors from the VICTORIA trial,
TES-REDUCED trial randomized 456 clinically stable patients however, it is unclear how the combination of vasodilatory
with chronic heart failure recently hospitalized for HF and therapies would be tolerated or would affect clinical outcomes
with LVEFs of less than 45% to either placebo or one of four (Armstrong 2020). Therefore, the traditional, vasodilatory com-
oral vericiguat doses ranging from 1.25 to 10 mg daily for 12 bination of isosorbide dinitrate and hydralazine still remains
weeks. Although vericiguat was well tolerated, the primary strongly preferred to vericiguat for African American patients
end point of change in NT-proBNP was no different from that with HFrEF as a compulsory add-on to GDMT (Maddox 2021).
of placebo. The phase III Vericiguat Global Study in Subjects
with Heart Failure with Reduced Ejection Fraction (VICTORIA) OPTIMIZING GDMT
trial, however, achieved its primary composite end point of For decades, the original, evidence-based pillars of GDMT
cardiovascular death or first HHF (Armstrong 2020). In the for HFrEF have included RAAS inhibitors and sympathetic
VICTORIA study, 5050 patients with NYHA II–IV HF and LVEFs nervous system antagonists from three primary pharmaco-
of less than 45% on optimal medical therapy were randomized logic categories: angiotensin-converting-enzyme inhibitors
to receive vericiguat to a target 10 mg or placebo. Driven pri- (ACEIs) or angiotensin receptor blockers (ARBs), β-block-
marily by a reduction in HHF, vericiguat modestly reduced the ers, and MRA. Despite long-standing availability, widespread
risk of the primary outcome (37.9% vs. 40.9%; HR 0.90; 95% CI, affordability, and robust evidence to support the strongest
0.83–0.98; p=0.02) after a median 10.8 months of follow-up. possible guideline recommendations for use in all patients
Neither symptomatic hypotension nor syncope occurs more with stage C HFrEF, less than 25% of eligible patients are pre-
often with vericiguat. scribed all three medication classes in contemporary practice
In light of guideline preference for ARNI and the addition (Greene 2018). Furthermore, a dismal 1% of those patients are
of SGLT2 inhibitors to GDMT, vericiguat’s place in therapy simultaneously titrated to target doses proven to extend sur-
should be reserved for patients with advanced HFrEF and vival in pivotal clinical trials (Table 3).

Table 3. Evidence-Based Dosing Guidance for HFrEF Medications

Mean Total Daily Dose


Starting Dose Target Dose Achieved in Clinical Trials

ACEI (Class I; LOE A)

Captopril 6.25 mg TID 50 mg TID 122.7 mg

Enalapril 2.5 mg BID 10–20 mg BID 16.6 mg

Lisinopril 2.5–5 mg 20–40 mg 32.5–35 mg

Ramipril 1.25–2.5 mg 10 mg 7.7 mg

Trandolapril 0.5–1 mg 4 mg 2.5 mg

ARB (Class I; LOE A)

Candesartan 4–8 mg 32 mg 24 mg

Losartan 50 mg 150 mg 129 mg

Valsartan 40 mg BID 160 mg BID 254 mg

ARNI (Class I; LOE B)

Sacubitril/Valsartan 24/26–49/51 mg 97/103 mg BID 375 mga

(continued)

PSAP 2022 Book 1 • Cardiology 15 Heart Failure with Reduced Ejection Fraction
Table 3. Evidence-Based Dosing Guidance for HFrEF Medications  (continued)

Mean Total Daily Dose


Starting Dose Target Dose Achieved in Clinical Trials

β-Blockers (Class I; LOE A)

Bisoprolol 1.25 mg 10 mg 8.6 mg

Carvedilol 3.125 mg BID 25 mg BID for weight <85 kg 37 mg


50 mg BID for weight ≥85 kg

Carvedilol CR 10 mg 80 mg Not studied

Metoprolol succinate 12.5–25 mg 200 mg 159 mg

MRA (Class I; LOE A)

Eplerenone 25 mg QD 50 mg 42.6 mg

Spironolactone 12.5–25 mg 25-50 mg 26 mg

sGC Modulatorsb

Vericiguat 2.5 mg QD 10 mg 8.9 mg

SGLT2 Inhibitor b

Dapagliflozin 10 mg 10 mg Not reported

Empagliflozin 10 mg 10 mg Not reported

Vasodilators (Class I; LOE A)

Hydralazine 25 mg TID 75 mg TID Not studied

Isosorbide dinitrate 20 mg TID 40 mg TID Not studied

Fixed-dose combination hydralazine/ 20/37.5 mg TID 40/75 mg TID 90 mg/175 mg


isosorbide dinitratec

If Channel Inhibitor (Class IIA; LOE B)

Ivabradine 2.5–5 mg BID Titrate to HR 50–60 bpm 13 mg


Maximum dose 7.5 mg BID

Total daily dose of both individual sacubitril/valsartan components described in the PARADIGM-HF trial.
a

b
Both sGC modulators and SGLT2I became recently approved for HFrEF, but only SGLT2Is are endorsed in updated Expert
Consensus Decision Pathways as GDMTs.
c
The ACC/AHA/HFSA guideline considers either the fixed-dose combination or the separate combination of isosorbide dinitrate and
hydralazine appropriate for GDMT for HFrEF in self-described Black patients, but it is important to note that these
recommendations do not include isosorbide mononitrate.
ACC = American College of Cardiology; ACEI = angiotensin-converting-enzyme inhibitor; AHA = American Heart Association;
ARB = angiotensin receptor blocker; ARNI = angiotensin receptor–neprilysin inhibitor; BID = twice daily; bpm = beats per minute;
GDMT = guideline-directed medical therapy; HFrEF = heart failure with reduced ejection fraction; HFSA = Heart Failure Society of
America; HR = heart rate; LOE = level of evidence; MRA = mineralocorticoid receptor antagonist; QD = once daily; sGC = soluble
guanylate cyclase; SGLT2i = sodium-glucose cotransporter-2 inhibitor.
Information from: Maddox TM, Januzzi JL, Allen LA, et al. 2021 Update to the 2017 ACC Expert Consensus Decision Pathway for
Optimization of Heart Failure Treatment: Answers to 10 Pivotal Issues About Heart Failure With Reduced Ejection Fraction:
A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 2021;77:772-810.

Because ARNI preference and SGLT2 inhibitor incorpo- patients without diabetes presumably even lower (Vadugana-
ration into GDMT became endorsed only more recently by than 2020). Plus, positioning SGLT2 inhibitors as the fourth
guidelines and expert consensus statements, adoption of pillar of GDMT for HFrEF management may reduce cumula-
these newer therapies represents an immediately impact- tive mortality risk by 73%, thereby preventing one death for
ful quality improvement opportunity. For example, just 2% every 3.9 patients treated over 2 years (Bassi 2020). All four
of patients with HFrEF and comorbid T2DM are currently disease-modifying therapeutic classes have early, incremen-
treated with SGLT2 inhibitors—with prescribing rates for tal, and additive benefits when used as quadruple therapy,

PSAP 2022 Book 1 • Cardiology 16 Heart Failure with Reduced Ejection Fraction
yet they remain woefully underutilized at present for a multi- maximally tolerated. Subtarget dosing still confers a magni-
tude of reasons. tude of benefit—precluding the need to achieve target doses
Barriers to optimal medication titration are indeed multi­ before initiating other disease-modifying therapy. Previous
faceted but most often include patient-specific factors intolerance should not obviate future titration attempts in the
such as abnormal kidney function, hyperkalemia, hypoten- absence of contraindications. Furthermore, tolerability may
sion, pill burden, or out-of-pocket expenses—particularly in be improved by initiating GDMT at lower starting doses or by
the incorporation of more-recently-approved therapies such staggering individual agents—particularly ARNI and β-block-
as ARNI and SGLT2 inhibitors (Maddox 2021). Provider- and ers (Greene 2021). Simultaneous initiation of MRA and SGLT2
health-system-related impediments to the implementation of inhibitors, however, requires no or minimal titration and rarely
established GDMT may also play a role. Clinical inertia, siloed precipitates adverse effects.
care, poor interprovider communication, and lack of familiar- Recent guidelines endorse ARNI initiation to be concurrent
ity with highly complex HF regimens are among the chief con- with β-blocker therapy, followed by adding MRA and SGLT2
tributing nonpatient factors. Therefore, routine integration of inhibitors 2–4 weeks later (Maddox 2021). With continual
pharmacists into collaborative, multidisciplinary-care path- potassium, creatinine, and vital sign monitoring, doses of
ways dedicated to HF management are critical to optimizing each medication can be increased on a biweekly basis until
GDMT and promoting medication adherence. goal or maximal tolerated dose is achieved. More-intensive
approaches to GDMT optimization support synchronized ini-
Sequence of Initiation and Titration tiation of all four foundational therapies at once, completing
The fundamental principle of HF care is to prioritize the titration to target doses as soon as 3 weeks (Greene 2021).
use of GDMT to derive the greatest potential benefit. For Alternative sequencing strategies propose simultaneous
all patients with HFrEF, this now includes quadruple ther- initiation of β-blockers and SGLT2 inhibitors first, followed
apy with ARNI, evidence-based β-blockers, MRA, and SGLT2 by ARNI and MRA within 4 weeks thereafter (McMurray
inhibitors (Maddox 2021). In addition, the combination of 2021). Uniquely, uptitration to target doses with this model
hydralazine and isosorbide dinitrate is also first-line therapy occurs only after quadruple therapy has been established.
for all self-identified African Americans, and ivabradine may A thorough understanding of medication-specific factors—
be considered second-line for select HFrEF patients in sinus including mechanism of action, common adverse effects,
rhythm (Figure 1). and routine monitoring parameters—is key to successful
Conventional sequencing of GDMT was cumbersome and titration of GDMT to target dosing (Table 4). Irrespective of
protracted, typically requiring at least 6 months to reach tar- the particular pathway chosen, an individualized approach
get doses while also necessitating a trial period of ACEI or ARB that prioritizes both the timely initiation and the optimization
before transitioning to ARNI (Yancy 2017). Although more- of GDMT must also coincide with close clinical assessment
recent guideline recommendations endorse preference for and consideration of patient-specific barriers to medication
ARNI initiation in de novo HFrEF, transitioning stable patients titration.
on ACEIs or ARBs is still a commonly encountered clini-
cal scenario. Patients on equivalent ACEIs that total a daily
dose of more than 10 mg of enalapril can begin sacubitril/ INOTROPIC THERAPY
valsartan 49/51 mg twice daily after a 36-hour washout The term inotropic therapy refers broadly to pharmacologic
period. The same dose of sacubitril/valsartan is also recom- treatments that improve cardiac contractility. Although clin-
mended for patients on an equivalent total daily dose of more ically useful for acute treatment of cardiogenic shock with
than 160 mg of valsartan. Patients on fewer ACEI/ARB equiva- low cardiac output, inotropes are associated with adverse
lents and who have eGFRs of less than 30 mL/minute/1.73 m2 outcomes when used for chronic HF. Despite a multitude
are recommended for initiation with sacubitril/valsartan of attempts, short-term improvements in hemodynamics or
24/26 mg twice daily. surrogate measures of cardiac performance with inotropic
Within 1 or 2 weeks of initiation, GDMT begins to reduce agents have not translated into longer-term morbidity bene-
the risk of cardiovascular death, HHF, and urgent HF visits, fits and have even increased mortality (Ahmad 2019). How-
and based on that, treatment should begin immediately in ever, initial investigation of inotropic therapy was conducted
stable patients (Packer 2021). Deferring optimal medical ther- before incorporation of β-blockers and cardiac resynchro-
apy reduces the likelihood of ever initiating GDMT and there- nization therapy or implantable cardioverter-defibrillator
fore may also result in preventable harm. Opportunities to devices (CRT/ICD) into routine practice. Given the growing
initiate and uptitrate GDMT in the hospital setting or during population of patients with end-stage HF, the high cost of
the vulnerable, postdischarge period should not be missed. ventricular-assist devices, and the organ shortage facing car-
Furthermore, because target doses of each medication are diac transplantation, interest in the development of novel and
associated with the best possible outcomes, titration to safe inotropes possessing unique mechanisms of action has
these dosing thresholds should be attempted until reached or been reinvigorated after a substantial hiatus.

PSAP 2022 Book 1 • Cardiology 17 Heart Failure with Reduced Ejection Fraction
At risk of HF (Stage A) Pre-HF (Stage B) Disease Prevention

• Risk Factor Management • Manage Structural Cardiac Disease


• Diabetes • MI → BB, DAPT, Statin
• Hypertension • MI with EF ≤35% → ACEI/ARB, BB, DAPT, MRA, Statin
• Hyperlipidemia • NICM → ACEI/ARB, BB

HF (Stage C) Initiate/Optimize GDMT

Comorbidity
• Elevated BNP or recent HHF → ARNI, MRA
HFpEF management,
• HTN → ACEI/ARB
Diuretics, SGLT2Ia

NYHA II-IV
• eGFR ≥20–30 → SGLT2Id
• eGFR ≥30 & K ≤5 → MRA

NYHA I–IV (EF ≤40%)


NYHA III or IV
HFrEF ACEI/ARB/ARNIb
• AA on GDMT → HYDISDN
AND BBc

NYHA II or III (EF ≥35%)


• HR ≥70, max BB → Ivabradine

Advanced HF (Stage D) Durable MCS, Palliation, Symptom Control

• Cardiac transplantation
• Inotropic therapy
• LVAD implantation

Figure 1. Treatment algorithm for HF.


a
Empagliflozin reduced the risk of cardiovascular death or HHF among patients with HFpEF.
b
ARNI is preferred to ACEI or ARB as first-line therapy. Prior ACEI/ARB tolerance is not required.
c
Evidence-based β-blockers include bisoprolol, carvedilol, and metoprolol succinate. Though also recommended, carvedilol CR was
approved on the basis of pharmacokinetic equivalence.
d
Empagliflozin if eGFR is 20–29 or more mL/minute/1.73 m2. Either dapagliflozin or empagliflozin if eGFR is 30 or more mL/minute/1.73 m2.
AA = African American; ACEI = angiotensin-converting-enzyme inhibitor; ARB = angiotensin receptor blocker; ARNI = angiotensin
receptor–neprilysin inhibitor; BB = β-blocker; CRT = cardiac resynchronization therapy; DAPT = dual antiplatelet therapy; EF = ejection
fraction; eGFR = estimated glomerular filtration rate; GDMT = guideline-directed medical therapy; HF = heart failure; HFpEF = heart
failure with preserved ejection fraction; HFrEF = heart failure with reduced ejection fraction; HHF = hospitalization for heart failure;
HR = heart rate; HTN = hypertension; HYD-ISDN = hydralazine-isosorbide dinitrate; ICD = implantable cardioverter-defibrillator;
K = potassium; MCS = mechanical circulatory support; MI = myocardial infarction; MRA = mineralocorticoid receptor antagonist;
NICM = nonischemic cardiomyopathy; NYHA = New York Heart Association; SGLT2I = sodium-glucose cotransporter 2 inhibitor.
Information from: Maddox TM, Januzzi JL, Allen LA, et al. 2021 Update to the 2017 ACC Expert Consensus Decision Pathway for
Optimization of Heart Failure Treatment: Answers to 10 Pivotal Issues About Heart Failure With Reduced Ejection Fraction:
A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 2021;77:772-810.

PSAP 2022 Book 1 • Cardiology 18 Heart Failure with Reduced Ejection Fraction
Table 4. HF Therapies: Mechanism of Action, Adverse Effects, and Monitoring

Drug Class Mechanism of Action Adverse Effects Monitoring Parameters

ACEI Inhibit ACE, reducing Ang I conversion to Ang II • Acute kidney injury • BP
• Angioedema • K
• Cough • SCr
• Hyperkalemia
• Hypotension

ARB Antagonize Ang II type 1 receptor • Acute kidney injury • BP


• Angioedema • K
• Hyperkalemia • SCr
• Hypotension

ARNI Inhibit neprilysin, potentiating natriuretic peptide • Acute kidney injury • BP


activity Antagonize Ang II type 1 receptor • Angioedema • K
• Cough • NT-proBNP
• Hyperkalemia • SCr
• Hypotension

β-Blockers Antagonize β-adrenergic receptor and potentially • Bradycardia • BP


α-adrenergic activity • Dizziness • HR
• Hypotension

Digoxin Inhibit Na/K ATPase pump, increasing intracellular • Bradycardia • BP


Na and Ca influx, improving impaired baroreceptor • Confusion • Digoxin level
reflex and renal Na reabsorption, and inhibiting • Dizziness • HR
sympathetic activity • Nausea
• Visual disturbances
• Vomiting

Hydralazine Direct arteriole vasodilation • Dizziness • BP


• Headache
• Hypotension

If Channel Selective sinoatrial node funny-channel-current- • Bradycardia • BP


Inhibitors flow disruption • Hypotension • HR
• Phosphenes

Isosorbide Increase cGMP concentration, relaxing vascular • Dizziness • BP


dinitrate smooth muscle • Headache
• Hypotension

Loop Diuretics Inhibit Na reabsorption in ascending loop of Henle • Dizziness • BP


• Continual urination • Urine output
• Hypotension • Volume status
• Muscle cramps • Weight
• Thirst

MRA Antagonize mineralocorticoid receptor, blocking • Gynecomastia • BP


aldosterone (spironolactone) • K
• Hyperkalemia • SCr

sGC Modulators Increase cGMP concentration, relaxing vascular • Anemia • BP


smooth muscle • Hypotension • CBC

SGLT2I Antagonize Na-glucose cotransporter in proximal • Acute kidney injury • A1C


tubule, reducing renal glucose reabsoprtion • Dyslipidemia • BP
• Genital mycotic • Glucose
infection • SCr
• Hypoglycemia • Volume status
• Ketoacidosis
• Urinary tract infection

(continued)

PSAP 2022 Book 1 • Cardiology 19 Heart Failure with Reduced Ejection Fraction
Table 4. HF Therapies: Mechanism of Action, Adverse Effects, and Monitoring  (continued)

ACEI = angiotensin-converting enzyme inhibitor; Ang I = angiotensin I; Ang II = angiotensin II; ARB = angiotensin receptor blocker;
ARNI = angiotensin receptor–neprilysin inhibitor; BP = blood pressure; Ca = calcium; CBC = complete blood count; HF = heart failure;
HR = heart rate; K = potassium; MRA = mineralocorticoid receptor antagonist; Na = sodium; NT-proBNP = N terminal fragment B-type
natriuretic peptide; SCr = serum creatinine; sGC = soluble guanylate cyclase; SGLT2I = sodium-glucose cotransporter-2 inhibitor.

Because of both lack of demonstrated benefit and potential at lower doses but exerts a largely neutral impact on vascular
for harm, the current role of conventional inotropic agents— tone at higher doses because of a counterbalance of α1 and
chiefly dobutamine, and milrinone—is limited to patients with β2 receptor affinity. Although milrinone provides more-bal-
advanced HF refractory to GDMT for either palliation or as anced inodilation across the dosing spectrum, it is eliminated
bridge therapy to mechanical circulatory support or cardiac renally, and given its comparably longer half-life, it may accu-
transplant in eligible patients (Yancy 2013). Dobutamine—a mulate in kidney injury. Milrinone does avoid direct adrener-
β-agonist—and milrinone—a phosphodiesterase-3 inhibitor—­ gic agonism, which may be advantageous for patients with
directly improve cardiac contractility and provide varying HF-prescribed concomitant β-blocker therapy. Both agents
degrees of vasodilation through activity in the peripheral vas- are highly arrhythmogenic, however, which necessitates
culature. Dobutamine reduces systemic vascular resistance

Patient Care Scenario


A 56-year-old African American woman with a medical his- During a visit to the HF disease management clinic
tory significant for T2DM (A1C = 7.4% on metformin), CKD 2 weeks after discharge, she reports somewhat tolerating
stage 3 (baseline SCr 1.3; eGFR 45 mL/minute/1.73 m2), the new medication regimen aside from a few headaches,
and HFrEF (LVEF = 33%) was hospitalized for shortness fatigue, and episodes of lightheadedness. A basic met-
of breath and fluid overload consistent with an acute abolic panel from the clinic is unremarkable. Her HR is
decompensation requiring intravenous diuretics. During 72 beats/minute, and blood pressure is 105/75 mm Hg.
the hospital stay, her home losartan 100 mg daily was A repeat echocardiogram revealed her LVEF was remain-
changed to sacubitril/valsartan 97/103 mg twice daily; ing around baseline at 34%. What is best to recommend
home metoprolol succinate continued at 200 mg daily; for this patient?
and she was newly started on spironolactone 50 mg daily.

ANSWER
The sequencing of initiation and uptitration of GDMT often causes headache and dizziness but reduces sys-
for HFrEF is a common but complex clinical problem tolic and diastolic blood pressure by only 1.9 and 2.4
facing contemporary HF management. Before SGLT2 mm Hg, respectively; SGLT2 inhibitors reduce systolic
inhibitors’ demonstrations of cardiovascular mortal- blood pressure by about the same magnitude or less.
ity and HHF benefit when added on to ACEI/ARB/ARNI, Given her concomitant CKD and T2DM with an A1C above
β-blocker, and MRA, the combination of hydralazine and goal, there are compelling indications for initiation of an
isosorbide dinitrate would be indicated as part of GDMT SGLT2 inhibitor such as dapagliflozin or empagliflozin to
for self-identified Black patients given the 43% rela- reduce the risk of cardiorenal complications and thereby
tive reduction in mortality observed in the A-HeFT trial. preserve kidney function. To date, no heterogeneity by
Current guidance recommends African American patients ethnicity or race has been identified in SGLT2 inhibitor
with HFrEF be prescribed this combination after GDMT studies. Prioritizing the initiation of both the combina-
has been adjusted to target or maximally tolerated doses. tion of hydralazine and isosorbide dinitrate as well as the
This patient’s GDMT regimen was titrated to goal during SGLT2 inhibitor studied in HF would be consistent with
the hospital stay. Although her systolic blood pressure is the 2021 ACC Expert Consensus Decision Pathway and
only on the lower end of normal, she is already experienc- should be individualized to the patient’s comorbid condi-
ing side effects likely attributed to low blood pressure. tions and tolerance for additional GDMT that may further
The combination of hydralazine and isosorbide dinitrate lower blood pressure.

1. Maddox TM, Januzzi JL, Allen LA, et al. 2021 Update to the 2017 ACC Expert Consensus Decision Pathway for Optimization of Heart
Failure Treatment: Answers to 10 Pivotal Issues About Heart Failure With Reduced Ejection Fraction: A Report of the American College
of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 2021;77:772-810.
2. Taylor AL, Ziesche S, Yancy C, et al. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med
2004;351:2049-57.
3. Morris AA, Testani JM, Butler J. Sodium-glucose cotransporter-2 inhibitors in heart failure: racial differences and a potential for reduc-
ing disparities. Circulation 2021;143:2329-31.

PSAP 2022 Book 1 • Cardiology 20 Heart Failure with Reduced Ejection Fraction
careful monitoring and use of minimal effective doses to alle- driven by HHF and urgent-care visits; cardiovascular death
viate symptomatic congestion. alone was not affected (19.6% vs. 19.4%; HR 1.01; CI 0.92–
1.11). Potentially related to increased contractility, patients
Palliative HF Care with severely depressed LVEFs of 28% or less appeared to
Although successful implementation and titration of GDMT derive greater benefit than did patients with systolic dysfunc-
substantially improve survival, HF remains a debilitating and tions of more than 28%. A modest quality-of-life improvement
progressive disease with a poor overall prognosis. Therefore, in the Kansas City Cardiomyopathy Questionnaire was also
palliative care is strongly recommended for all patients with observed for patients recruited in the hospital but not in the
advanced HF in order to improve quality of life (Yancy 2013). ambulatory-care setting. Although NT-proBNP decreased by
Despite those recommendations and robust evidence for 10% at week 24, omecamtiv increased cardiac troponin levels
improved patient-centered health outcomes, palliative care 4 ng/L higher. Despite that finding, cardiac ischemic and ven-
is a persistently underutilized service that is rarely offered tricular arrhythmia events were similar to placebo.
to patients with HF (Diop 2017). A comprehensive palliative-­ Omecamtiv mecarbil awaits regulatory review, but its
care plan should be integrated early in HF management and place in therapy within the expanding HFrEF treatment
include intensive symptomatic relief, detailed end-of-life pref- landscape deserves further clarification. Based on GALAC-
erences, and access to caregiver support. Even in advanced TIC-HF, recently decompensated patients with poor systolic
HF, GDMT may be useful to extend life and avoid hospitaliza- functions may stand to benefit the most. Omecamtiv may
tion. Diuretic agents, however, are more critical to achieve
also be especially useful when hypotension or renal insuffi-
immediately control of symptoms of congestion and should
ciency limits the initiation or titration of GDMT as well as in
be continued through hospice to end of life (Maddox 2021).
patients with advanced or end-stage HF who may not be can-
Home infusions of dobutamine or milrinone should also be
didates for continuous inotrope infusions. Given that plasma
considered for palliation to improve functional status, though
concentration-guided dosing of omecamtiv mecarbil was
patients with end-stage HF may become (1) inotrope depen-
used in clinical trials, it is likely that pharmacists will play a
dent as their conditions worsen and (2) unable to wean with-
crucial role in its future use and monitoring.
out deterioration. As patients with HF transition toward
comfort care, discontinuation of life-sustaining inotropes
or neurohormonal antagonists may be prudent. These com- ADVANCED THERAPEUTIC
plex treatment decisions should be individualized to reflect a MODALITIES
patient’s wishes in concert with a palliative-care consultant. CRT/ICD Devices
Ventricular arrhythmias are common complications of HF
Investigational Therapies and are of concern. Arrhythmogenesis is often multifac-
torial because of increased sympathetic tone, underlying
Omecamtiv Mecarbil
ischemic heart disease or myocardial scarring, conduction
Omecamtiv mecarbil selectively activates cardiac myosin to
delays, electrolyte abnormalities, and drug-induced arrhyth-
promote conformational change of the physiologic, weakly
mias. Although GDMT may reverse cardiac remodeling and
bound myosin-ATP intermediate state to a stronger, actin-
lower arrhythmia risk, up to half of all patients with HF still
bound, force-producing state to stimulate myosin phosphate
release. This mechanism is thought to enhance the duration die from sudden cardiac death precipitated by a ventricular
and force of each systolic contraction without affecting myo- tachyarrhythmia (Virani 2021). Implantable cardiac devices
cardial energetics. Because omecamtiv acts directly on the consist of an electronic generator and lead wires, typically
sarcomere, it avoids certain potentially problematic calcium-­ placed by way of a minimally invasive procedure in a cardiac
related proarrhythmic effects observed with other inotro- catheterization or electrophysiology laboratory. Leads are
pes like digoxin, dobutamine, milrinone, and levosimendan inserted into a peripheral vein and guided toward the heart,
(Psotka 2019). one end attaching to the generator and the other terminat-
The Global Approach to Lowering Adverse Cardiac Out- ing in cardiac tissue itself. The generator is powered by lith-
comes Through Improving Contractility in Heart Failure ium batteries, which can last up to 10 years before requiring
(GALACTIC-HF) trial recently compared pharmacokinetically replacement, at which time the device must be accessed
guided dosing of omecamtiv mecarbil with placebo in 8256 from a tunneled pocket under the skin of the chest well.
patients with symptomatic chronic HF and LVEFs of 35% or Device pocket hematomas are rare but major risks of mor-
less (Teerlink 2021). After a median follow-up period of 21.8 bidity, necessitating cautious periprocedural anticoagulation
months, the primary composite outcome of HHF, urgent HF management. Subcutaneous systems are alternatives to the
visit, or cardiovascular death occurred significantly less traditional transvenous approach, featuring an entirely extra-
often with omecamtiv (37.0% vs. 39.1%; HR 0.92; CI 0.86– cardiac implantation procedure that avoids many of the com-
0.99; p=0.03). It is important to note that that difference was mon perioperative complications of conventional devices.

PSAP 2022 Book 1 • Cardiology 21 Heart Failure with Reduced Ejection Fraction
Implantable cardioverter-defibrillators (ICDs) detect and defibrillate an arrhythmia with a view to restore normal sinus
terminate ventricular dysrhythmias by way of single- or rhythm. Amiodarone, lidocaine, and nondihydropyridine cal-
dual-chamber defibrillator leads capable of electronic-shock cium channel blockers can potentially raise the defibrillation
delivery. A biventricular ICD, also known as a cardiac resyn- threshold and thereby require higher voltage or risk defibril-
chronization therapy (CRT) device, consists of a right-ven- lation failure and subsequent shocks. Contrastingly, β-block-
tricular defibrillator lead as well as an added, left-ventricular ers as well as sotalol or dofetilide may lower the defibrillation
pacing lead. The CRT devices optimize the atrioventricular threshold. These agents can also slow the rate of ventricu-
interval through the coronary sinus, thereby coordinating lar tachycardia to below rate-sensing cutoff programming,
contraction between and within both ventricles. Implantation thereby misfiring an appropriate ICD shock opportunity
of either cardiac device has been shown to improve outcomes (Lampert 2013). Amiodarone plus β-blockers appears to be
among patients with HFrEF for both primary and secondary more effective than sotalol to prevent device shocks—but at
arrhythmia prevention. Primary-prevention ICD placement the expense of more-adverse pulmonary and thyroid events
should be considered for patients with HFrEF and persistently as well as bradycardia (Connolly 2006). Drug–device inter-
reduced LVEFs of 35% or less despite at least three optimally actions represent an important component of CRT/ICD man-
dosed GDMTs (see Figure 1). For this indication, ICD therapy agement, and pharmacists are uniquely qualified and well
reduced all-cause mortality by more than 50% during a 5-year positioned to inform providers and patients so as to avoid
study period compared with conventional medical therapy in adverse events and improve quality of life.
patients with LVEFs of 30% or less, histories of myocardial
infarction, and inducible ventricular tachyarrhythmia (Moss Remote Hemodynamic Monitoring
1996). A subsequent study of 1232 similar patients recruited Development of remote patient monitoring accelerated
without requiring invasive electrophysiological testing found because of the 2019 coronavirus pandemic, thereby lever-
that defibrillator implantation reduced all-cause mortality by aging the accessibility and convenience of electronic, tele-
31% after only 20 months of average follow-up (Moss 2002). health, or mobile technology to manage and monitor disease
Among NYHA Class II or III patients with LVEFs of 35% or less outside traditional health care facilities. Remote patient
of either ischemic or nonischemic etiology, preventive ICD monitoring debuted in the area of HF almost 3 decades ago,
therapy reduced all-cause mortality by 23% compared with when a multidisciplinary telephonic monitoring and follow-­
amiodarone after a median 45.5 months of follow-up (Bardy up program reduced the rehospitalization rate by more
2005). Cardiac resynchronization therapy should be consid- than 50% (Rich 1995). However, subsequent large, prospec-
ered in symptomatic patients with EFs of 35% or less in sinus tive, multicenter investigations of more-advanced remote
rhythm with QRS durations of 120 msec or greater. Although patient-monitoring interventions—including implantable car-
CRT alone initially did not improve survival in 1520 patients diac electronic devices, thoracic bioimpedance or dielectric
with advanced HF and QRS intervals of at least 120 msec, sensing systems, and wearable hemodynamic sensors—have
the combination of CRT with a pacemaker-defibrillator did failed to demonstrate any benefit and were not cost-effective
reduce the risk of death by 36% compared with optimal phar- (Dickinson 2018). One notable exception is the CardioMEMS
macologic therapy (Bristow 2004). A subsequent CRT trial implantable pulmonary artery pressure-monitoring system,
did, however, demonstrate improved survival as well as echo- which received FDA approval in 2014 based on an analysis
cardiography findings, symptoms, and quality of life in 813 of long-term ongoing follow-up from the CardioMEMS Heart
patients with advanced HF because of systolic dysfunction Sensor Allows Monitoring of Pressure to Improve Outcomes
and cardiac dyssynchrony compared with standard pharma- in NYHA Class III Heart Failure Patients (CHAMPION) trial.
cologic therapy (Cleland 2005). Implantation of an implant- The CHAMPION trial was a prospective multicenter, single-
able cardioverter-defibrillator for secondary prevention has blind study of 550 patients with NYHA III HF who had had
also been shown to reduce mortality risk by 20%–30% and is recent HHFs in the past year and were randomized to receive
indicated for survivors of sudden cardiac arrest or those with wireless implantable pulmonary artery pressure monitors
syncopes from or histories of presumed sustained ventricu- compared with usual care (Abraham 2011). After a mean of 15
lar arrhythmia. It is important to note that patients at NYHA months follow-up, the CardioMEMS device had reduced the
stage IV have not been shown to benefit from ICD placement primary end point of HHF by 37% (158 vs. 254; HR 0.763; 95%
and should be considered only if awaiting heart transplanta- CI, 0.52–0.77; p<0.0001). Because HF decompensation is typ-
tion. In addition, to avoid inappropriate shocks that may be ically preceded by days to weeks of asymptomatic intracar-
inconsistent with goals of care, clinicians must also disable diac and pulmonary artery pressure elevations, patients who
cardiac devices in patients with HF who are transitioning to were managed with hemodynamic data from the CardioMEMS
hospice care. device were more likely to have GDMT titration performed as
Antiarrhythmic medications variably affect the defibril- well as to receive real-time diuretic dose adjustments before
lation threshold of an ICD. Defibrillation threshold refers to symptoms of congestion developed. Adverse event rates
the lowest amount of energy necessary to successfully were consistent with those of right-heart catheterization

PSAP 2022 Book 1 • Cardiology 22 Heart Failure with Reduced Ejection Fraction
but better than other permanent implants such as CRT/ICD. impact on overall prognosis (Virani 2021). Traditionally
Based largely on reductions in HHF, CardioMEMS was asso- characterized by a crude categorization of LVEF of 50% or
ciated with a comprehensive health care cost reduction of more, HFpEF actually represents a heterogenous subpop-
$13,190 per year per patient implanted with the device (Desai ulation of HF with a multitude of distinct pathophysiologic
2017). A postapproval CardioMEMS single-arm observational phenotypes inherently more complex than classification by
study demonstrated even greater benefits in all-cause hos- diastolic dysfunction alone (Riello 2021). Impaired left ven-
pitalizations across HF subgroups, including patients with tricular relaxation is a common hallmark sign of HFpEF, but
HFpEF (Shavelle 2020). A pre-COVID-19-pandemic analysis of it may be caused by a host of interdependent factors such
the recent Haemodynamic-Guided Management of HF study as cardiometabolic disease and systemic inflammation, obe-
suggested a possible benefit on mortality and total HF events sity and epicardial adipose accumulation, myocardial isch-
based on hemodynamic-guided management through the emia and fibrosis, and arterial and vascular rigidity (Obokata
CardioMEMS monitoring system (Lindenfeld 2021). Pharma- 2020). Although active development of disease-modifying
cists participating in remote hemodynamic-monitoring pro- pharmacotherapy for HFpEF has remained unsuccessful for
grams may be able to optimize GDMT or guide diuretic dose several decades, recent emerging research and regulatory
titrations to prevent HHF and reduce total cost of care. expansion suggest that this area of tremendous need may
soon establish more-definitive treatment—and with measur-
LVAD and Cardiac Transplantation able benefit beyond symptomatic control and comorbidity
Patients with advanced HF refractory to GDMT should be management.
considered for advanced therapeutic modalities such as
definitive therapy, including durable mechanical circulatory Angiotensin- Receptor–Neprilysin-Inhibitor
support with left ventricular assist devices (LVADs) or refer- Given its landmark success in HFrEF and encouraging mul-
rals for orthotopic heart transplantation (see Figure 1). Car- tifactorial impact on several compensatory neurohormonal
diac transplantation is the only curative treatment for HF pathways thought to be at least partially shared with HFpEF,
and has a median donor graft survival of more than 12 years. patients, clinicians, and investigators were hopeful that ARNI
However, the paucity of suitable organ donors renders heart may be among the first medications to show a clear and con-
transplantation an epidemiologically insignificant therapeu- sistent benefit in HFpEF. The Prospective Comparison of
tic strategy when compared with the growing need of poten- ARNI with ARB Global Outcomes in HF with Preserved Ejec-
tial stage D recipients. Therefore, to extend life with improved tion Fraction (PARAGON-HF) study randomized 4796 symp-
functional status, the implantation of an LVAD may be use- tomatic patients with LVEFs of 50% or more and elevated
ful as a bridge to transplantation or used as destination ther- NT-proBNP to sacubitril/valsartan 97/103 mg twice daily or
apy in those not eligible for transplantation. End-of-life goals valsartan 160 mg twice daily (Solomon 2019). Although ARNI
and individualized, patient-specific characteristics should be reduced the primary composite end point of cardiovascular
reviewed thoroughly in consideration of advanced therapeu- death and HHF by 13%, it narrowly missed statistical signifi-
tic modalities. For example, pulmonary hypertension is a sig- cance after a median 35 months of follow-up (894 vs. 1009;
nificant contraindication to transplantation but not for LVAD HR 0.87; 95% CI, 0.75–1.01; p=0.059). The fragility index of this
candidacy. Conversely, patients with severe right ventricular result required a net difference of only seven events to reach
failure are suboptimal LVAD candidates but may experience a p value of less than 0.05 (Solomon 2021). Furthermore, had
better transplantation outcomes. Other LVAD precautions urgent HF visits—which were collected prospectively and
include history of recurrent infections, untreated aortic regur- adjudicated independently—been incorporated into the pri-
gitation, older age, and high frailty index. Anticoagulation with mary composite outcome, as with other contemporary HFpEF
warfarin to an INR goal of 2–3 is currently recommended to trials, PARAGON-HF would have indeed achieved its end point
prevent LVAD thrombosis for all available devices. Alternative (HR 0.86; 95% CI, 0.75–0.99; p=0.040). There was significant
anticoagulation strategies with newer-generation VADs that heterogeneity of the trial findings between two prespecified
have magnetically suspended propulsion systems are cur- subgroups—including hypothesis-generating observations
rently under investigation, potentially obviating the need for of more-favorable effects with ARNI in those with LVEFs of
anticoagulation altogether, which may significantly reduce 45% to 57%. as well as in women compared with men. Nota-
complications if pump thrombosis risk can be minimized. bly, ARNI was also associated with an improvement in the
exploratory renal composite outcome of death from kid-
HEART FAILURE WITH PRESERVED ney failure, end-stage renal disease, and an eGFR decrease
EJECTION FRACTION of 50% or more from baseline (1.4% vs. 2.7%; HR 0.50; 95%
Despite accounting for approximately half of the overall HF CI, 0.33–0.77). It was important that fewer patients random-
population and conferring a comparably high mortality risk, ized to sacubitril/valsartan also discontinued the study drug
HFpEF has historically lacked even a single evidence-based and had SCr elevations of 2.0 or more mg/dL or any elevated
pharmacologic treatment option offering an observable

PSAP 2022 Book 1 • Cardiology 23 Heart Failure with Reduced Ejection Fraction
serum potassium but did experience more hypotension and recommend consideration of an MRA for patients with HFpEF
angioedema. and LVEFs or more than 45%, elevated BNPs, and recent hos-
In consideration of the totality of evidence—particu- pitalizations within the past year to reduce the risk of sub-
larly across the spectrum of LVEF consistent with PARA- sequent HHF (see Figure 1). Much like the recent ARNI label
DIGM-HF—the FDA Cardiovascular and Renal Drugs Advisory expansion, the same FDA Cardiovascular and Renal Drugs
Committee voted to approve an expanded ARNI indication “to Advisory Committee also voted that the totality of evidence
reduce the risk of cardiovascular death and hospitalization from TOPCAT was compelling enough to support a broader
for heart failure in patients with chronic heart failure,” with indication for spironolactone inclusive of at least patients
no specific LVEF cutoff (Solomon 2021). Although the merits with HFmrEF up to LVEFs of 55% – 57%. However, a formal
of the largely unprecedented regulatory decision to expand label expansion request has yet to be submitted to the FDA
ARNI labeling to support use in HFpEF despite a neutral trial for consideration.
finding are contentious, it may be reasonable to consider Another recently approved MRA, finerenone, is currently
sacubitril/valsartan for female patients with HFpEF as well being investigated in the ongoing FINEARTS-HF trial for a
as those with EFs of 57% or less who are at low risk of symp- potential impact on the primary composite end point of car-
tomatic hypotension. diovascular death, HHF, or urgent HF visits in HFpEF (clini-
caltrials.gov). About 5550 patients with LVEFs of 40% or
Mineralocorticoid Receptor Activation more, NYHA II–IV symptoms, elevated NT-proBNPs, struc-
Mineralocorticoid receptor activation is associated with tural heart disease, and recent HF events will be randomized
sodium retention, potassium loss, endothelial dysfunction, in either an ambulatory or an acute-care setting to receive
vascular inflammation, myocardial fibrosis, and hypotrophy finerenone 40 mg once daily or matching placebo. Should
central to the pathophysiology of HFrEF and is shared, at finerenone demonstrate an impact similar to that in the FIDE-
least in part, by HFpEF. Therefore, the use of MRA to manage LIO-DKD and FIGARO-DKD trials in the area of cardiorenal end
diastolic dysfunction in HFpEF was once a promising ther- points among patients with nondiabetic HFpEF, it may be the
apeutic target that initially disappointed when studied rig- first MRA to definitively mitigate morbidity and mortality risk
orously. The Treatment of Preserved Cardiac Function with beyond HFrEF.
an Aldosterone Antagonist Trial (TOPCAT) was a large mul-
ticenter, double-blind, placebo-controlled trial that evalu- SGLT2 Inhibitors
ated the effect of spironolactone on morbidity and mortality Sodium-glucose cotransporter 2 inhibitors have consistently
in 3445 patients with HFpEF (Pitt 2014). The primary com- demonstrated cardiorenal outcome benefits among patients
posite end point of cardiovascular death, HHF, and resusci- with HFrEF and patients with CKD independent of T2DM. Nota-
tated cardiac arrest was similar between treatment arms bly, several pivotal cardiovascular safety studies of patients
(18.6% vs. 20.4%; HR 0.89; 95% CI, 0.77–1.04; p=0.14) after with T2DM at high cardiovascular risk—such as EMPA-REG,
a mean follow-up of 3.3 years, as were cardiovascular mor- CANVAS, and DECLARE-TIMI 58—did not distinguish comor-
tality and aborted cardiac arrest individually. Rates of HHF, bid HF subpopulations by ejection fraction. Cardiovascu-
however, were significantly lower with spironolactone (12.0% lar outcome trials with pretrial ejection fraction information
vs. 14.2%; HR 0.83; 95% CI, 0.69–0.99; p=0.04). Furthermore, available like VERTIS CV, SCORED, and SOLOIST suggest
hyperkalemia and renal failure were also more common in a benefit of HHF among patients with comorbid T2DM and
spironolactone-treated patients. Interestingly, a post hoc HFpEF but remain unproven in nondiabetic HFpEF. Given that
analysis of TOPCAT revealed significant geographic dis- diabetic cardiomyopathy can manifest as either systolic or
parities with regard to the primary outcome between study diastolic function, independent trials were necessary to con-
sites in North America and South America (27.3% vs. 31.8%; firm the hypothesis-generating benefits of SGLT2 inhibitors
HR 0.82; 95% CI, 0.69–0.98; p=0.26) compared with those in for both HFrEF and HFpEF. Based on the landmark findings of
eastern Europe (9.3% vs. 8.4%; HR 1.10; 95% CI, 0.79–1.51; DAPA-HF and EMPEROR-Reduced, dapagliflozin and empagli-
p=0.576) (Pfeffer 2015). About half of all patients enrolled in flozin are now considered part of quadruple GDMT for HFrEF;
TOPCAT were recruited from Russia and Georgia, experienc- however, only empagliflozin has yet completed its HFpEF trial
ing curiously low event rates consistent with a healthier study to date (Maddox 2021).
population not necessarily suffering from HFpEF. In addition The Empagliflozin Outcome Trial in Patients with Chronic
to these regional biases, serum concentrations of the active Heart Failure (EMPEROR-Preserved) trial randomized 5988
spironolactone metabolite, canrenone, were undetectable patients with LVEFs of more than 40%, NYHA II–IV symptoms
in 30% of participants from Russia versus only 3% from the for 3 months or more before enrollment, elevated NT-proBNPs,
United States and Canada (de Denus 2017). These findings and either structural heart disease or recent HHF to empagli-
suggest, at a minimum, that lack of study drug adherence as flozin 10 mg once daily or placebo for a median 26.2 months
well as other potential protocol violations may have occurred of treatment. Empagliflozin significantly reduced the risk of
disproportionately outside the Americas. Current guidelines the primary composite end point of cardiovascular death or

PSAP 2022 Book 1 • Cardiology 24 Heart Failure with Reduced Ejection Fraction
HHF (13.8% vs. 17.1%; HR 0.79; 95% CI, 0.69–0.90; p<0.001),
Practice Points
driven predominantly by a 29% lower risk of hospitalization
(Anker 2021). Although there was a nonsignificant 9% lower Clinical pharmacists must overcome many barriers in
their optimizations of pharmacotherapies for patients
risk of cardiovascular death with empagliflozin (HR 0.91; 95%
with HF. As new strategies to better inform implemen-
CI, 0.76–1.09), overall mortality was neutral (HR 1.00; 95% CI, tation of GDMT for HFrEF and promising therapeutics
0.87–1.15). It is important to note that those benefits were for HFpEF loom on the horizon, new guideline rec-
consistent among patients with or without diabetes; however, ommendations as well as expanded indications for
an attenuation appeared to be observed in patients with the existing medications continuously evolve contempo-
highest ejection fractions (Anker 2021). Adverse events more rary practice:
commonly experienced with empagliflozin than with pla- • The HFSA/HFA-ESC/JHFS recently proposed a universal
cebo included uncomplicated UTI and hypotension. Although definition of HF and updated standardized HF classifi-
the high rate of treatment discontinuation (23%) was similar cations and staging beyond LVEF and structural cardiac
disease.
between arms, that discontinuation may have notably mini-
• Patients with HF should be routinely assessed and treated
mized the effect size of empagliflozin on cardiovascular and for common comorbidities such as iron deficiency, CKD,
all-cause mortalities (Drazner 2021). The Dapagliflozin Evalu- and T2DM to improve quality of life and reduce cardiorenal
ation to Improve the Lives of Patients with Preserved Ejection complications.
Fraction Heart Failure (DELIVER) trial is a similar interna- • Dapagliflozin and empagliflozin have recently been
shown to improve cardiovascular death rates and HF
tional, randomized, placebo-controlled HFpEF study eval-
hospitalizations—independent of diabetes status—among
uating the impact of dapagliflozin 10 mg once daily on the patients with HFrEF.
primary composite end point of cardiovascular death, HHF, • Clinicians should ensure that patients with HFrEF are
or urgent HF visits after about 39 months of follow-up (clini- receiving quadruple therapy with a RAAS inhibitor—
caltrials.gov). To date, the DELIVER study has recruited 6263 preferably, ARNI—in combination with a β-blocker, an MRA,
ambulatory or hospitalized patients with LVEFs greater than and now also an SGLT2 inhibitor.
• GDMT for HFrEF should be titrated to maximally tolerated
40%, evidence of structural heart disease, NYHA II–IV func-
doses from randomized, controlled trials but not at the
tional status for 6 weeks or more before enrollment, and ele- expense of initiating all four disease-modifying agents.
vated NT-proBNPs. It is anticipated that the results of both • Optimization of evidence-based therapy should occur every
trials will establish SGLT2 inhibitors as constituting the first 2 weeks during hospitalization for HFrEF and when patients
medication class to offer indisputable cardiovascular out- are outpatients so as to achieve GDMT within 3–6 months
of diagnosis.
come benefits among patients with HFpEF.
• Despite arguably neutral HFpEF trials, ARNI and spirono-
lactone recently received regulatory support for expanded
CONCLUSION
use in patients with LVEFs greater than 40%.
Four classes of disease-modifying therapeutics proven to • SGLT2 inhibitors may represent the first medication class
reduce cardiovascular mortality and prevent HF hospitaliza- in HFpEF to definitively improve cardiovascular mortality
tions are now available for patients with HFrEF. The medica- and HF hospitalizations.
tions—ARNIs, β-blockers, MRAs, and, most recently, SGLT2
inhibitors—are all supported by robust clinical trial evidence Although ARNI and MRA recently received regulatory support
as well as the strongest possible guideline recommendation for expanded indications in HFpEF, SGLT2 inhibitors have
compelling their use. Nonetheless, HF remains the costliest now become the only pharmacologic treatment options with
condition in the United States, with a 5-year mortality rate clear potential to significantly improve cardiorenal outcomes
comparable to most of the major malignancies. One of the for patients with HF, irrespective of ejection fraction or diabe-
most critical reasons for that is the abysmal uptake of evi- tes status. Given the pervasive underutilization of GDMT in
dence-based pharmacotherapy across the country. Recent patients with HFrEF, as new therapeutics emerge and prove
guidance on pathways for initiation, titration, and sequencing beneficial for HFpEF, pharmacists must ensure the same
of GDMT as well as how to address barriers to medication opti- latency to optimize that evidence-based treatment not also
mization provides a framework for closing that gap in care. affect HFpEF.
The integration of pharmacists into multidisciplinary care
teams dedicated to this endeavor represents a necessary step REFERENCES
toward optimal GDMT implementation, promotes best possi- Abraham WT, Adamson PB, Bourge RC, et al. Wireless
ble patient outcomes, and ensures medication adherence. pulmonary artery haemodynamic monitoring in chronic
Furthermore, recent recognition of the increasing prevalence heart failure: a randomised controlled trial. Lancet
and heterogeneity of HFpEF, without any definitive therapies 2011;377:658-66.
to mitigate its formidable morbidity and mortality, has rein- ADA (American Diabetes Association). 10. Cardiovascular
vigorated clinical investigators and regulatory agencies alike disease and risk management: Standards of Medical Care
to establish safe and effective therapies for this unmet need. in Diabetes 2021. Diabetes Care 2021 44:S125-S50.

PSAP 2022 Book 1 • Cardiology 25 Heart Failure with Reduced Ejection Fraction
Ahmad T, Miller PE, McCullough M, et al. Why has positive and improved clinical outcomes in HFrEF. J Am Coll
inotropy failed in chronic heart failure? Lessons learned Cardiol HF 2019;7:158-168.
from prior inotrope trials. Eur J Heart Fail 2019;21:1064-78.
de Denus S, O’Meara E, Desai AS, et al. Spironolactone
Anker S, Butler J, Filippatos G, et al. Empagliflozin in patients metabolites in TOPCAT: new insights into regional varia-
with chronic heart failure with preserved ejection fraction. tion. N Engl J Med 2017; 376:1690-2.
N Engl J Med 2021;doi:10.1056/NEJMoa2107038
Desai AS, Bhimaraj A, Bharmi R, et al. Ambulatory hemo-
Armstrong PW, Pieske B, Anstrom KJ, et al. Vericiguat in dynamic monitoring reduces heart failure hospitaliza-
patients with heart failure with reduced ejection fraction. tions in “real-world” clinical practice. J Am Coll Cardiol
N Engl J Med 2020;382:1883-93. 2017;69:2357-65.

Bakris GL, Agarwal R, Anker S, et al. Effect of finerenone Dickinson MG, Allen LA, Albert NA, et al. Remote monitoring
on chronic kidney disease outcomes in type 2 diabetes. of patients with heart failure: a white paper from the Heart
N Engl J Med 2020;383:2219-29. Failure Society of America Scientific Statements Commit-
tee. J Cardiac Fail 2018;24:682-94.
Bardy GH, Lee JL, Mark DB, et al. Amiodarone or an implant-
Diop MS, Rudolph JL, Zimmerman KM, et al. Palliative care
able cardioverter-defibrillator for congestive heart failure.
interventions for patients with heart failure: a systematic
N Engl J Med 2005;352:225-7.
review and meta-analysis. J Palliat Med 2017;20:84-92.
Bassi NS, Ziaeian B, Yancy CW, et al. Association of optimal
Drazner MH. SGLT2 inhibition in heart failure with a pre-
implementation of sodium-glucose cotransporter 2 inhib-
served ejection fraction – a win against a formidable foe.
itor therapy with outcome for patients with heart failure.
N Engl J Med 2021;doi:10.1056/NEJMed2113008
JAMA Cardiol 2020;5:948-51.
Dunlay SM, Givertz MM, Aguilar D, et al. Type 2 diabetes
Bhatt DL, Szarek M, Pitt B, et al. Sotagliflozin in patients mellitus and heart failure: a scientific statement from the
with diabetes and chronic kidney disease. N Engl J Med American Heart Association and the Heart Failure Society
2021a;384:129-39. of America. Circulation 2019;140:e294-e324.
Bhatt DL, Szarek M, Steg GP, et al. Sotagliflozin in patients Gerber Y, Weston SA, Redfield MM, et al. A contemporary
with diabetes and recent worsening heart failure. N Engl appraisal of the heart failure epidemic in Olmsted
J Med 2021b;384:117-28. County, Minnesota, 2000 to 2010. JAMA Intern Med 2015;
175:996–1004.
Bozkurt B, Coats AJS, Tsutsui H, et al. Universal definition
and classification of heart failure: a report of the Heart Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide
Failure Society of America, Heart Failure Association of and cardiovascular outcomes in type 2 diabetes (REWIND):
the European Society of Cardiology, Japanese Heart Fail- a double-blind, randomised placebo-controlled trial.
ure Society and Writing Committee of the Universal Defini- Lancet 2019;394:121-30.
tion of Heart Failure. J Card Fail 2021;27:P387-413.
Gheorghiade M, Greene SJ, Butler J, et al. Effect of vericig-
Bristow MR, Saxon LA, Boehmer J, et al. Cardiac-resynchro- uat, a soluble guanylate cyclase stimulator, on natriuretic
nization therapy with or without an implantable defibril- peptide levels in patients with worsening chronic heart
lator in advanced chronic heart failure. N Engl J Med failure and reduced ejection fraction: the SOCRATES-­
2004;350:2140-50. REDUCED randomized trial. JAMA 2015;314:2251-62.

Cannon CP, Pratley R, Dagogo-Jack S, et al. Cardiovascular Greene SJ, Butler J, Albert NM, et al. Medical therapy
outcomes with ertugliflozin in type 2 diabetes. N Engl J for heart failure with reduced ejection fraction: the
Med 2020;383:1425-35. CHAMP-HF registry. J Am Coll Cardiol 2018;72:351-66.

Cleland JGF, Daubert JCC, Erdmann E, et al. The effect of Greene SJ, Butler J, Fonarow GC. Simultaneous or rapid
cardiac resynchronization on morbidity and mortality in sequence initiation of quadruple medical therapy for heart
heart failure. N Engl J Med 2005;352:1539-49. failure—optimization therapy with the need for speed.
JAMA Cardiol 2021;6:743-4.
Connolly SJ, Dorian P, Roberts RS, et al. Comparison of
beta-blockers, amiodarone plus beta-blockers, or sotalol Heerspink HJL, Stefansson BV, Correa-Rotter R, et al.
for prevention of shocks from implantable cardioverter Dapagliflozin in patients with chronic kidney disease.
defibrillators: the OPTIC study: a randomized trial. N Engl J Med 2020;383:1436-46.
JAMA 2006;295:165-71. Heidenreich PA, Albert NM, Allen LA, et al; on behalf of the
American Heart Association Advocacy Coordinating Com-
Das SR, Everett BM, Birtcher KK, et al. 2020 Expert Consen-
mittee; Council on Arteriosclerosis, Thrombosis and Vas-
sus Decision Pathway on novel therapies for cardiovascu-
cular Biology; Council on Cardiovascular Radiology and
lar risk reduction in patients with type 2 diabetes: a report
Intervention; Council on Clinical Cardiology; Council on
of the American College of Cardiology Solution Set Over-
Epidemiology and Prevention; Stroke Council. Forecast-
sight Committee. J Am Coll Cardiol 2020;76:1117-45.
ing the impact of heart failure in the United States: a pol-
Daubert MA, Adams K, Yow E, et al. NT-proBNP goal achieve- icy statement from the American Heart Association. Circ
ment is associated with significant reverse remodelings Heart Fail 2013;6:606-19.

PSAP 2022 Book 1 • Cardiology 26 Heart Failure with Reduced Ejection Fraction
Hein AM, Scialla JJ, Edmonston D, et al. Medical manage- Obokata M, Reddy YNV, Borlaug BA. Diastolic dysfunction
ment of heart failure with reduced ejection fraction in and heart failure with preserved ejection fraction: under-
patients with advanced renal disease. JACC Heart Fail standing mechanisms by using noninvasive methods.
2019;7:371-82. JACC Cardiovasc Imagining 2020;13:245-57.

Husain M, Birkenfeld AL, Donsmark M, et al. Oral semaglu- Packer M, Anker SD, Butler J, et al. Cardiovascular and renal
tide and cardiovascular outcomes in patients with type 2 outcomes with empagliflozin in heart failure. N Engl J Med
diabetes. N Engl J Med 2019;381:841-51. 2020;383:1413-24.

Januzzi JL, Camacho A, Pina IL, et al. Reverse cardiac Packer M, Anker SD, Butler J, et al. Effect of empagliflozin
remodeling and outcome after initiation of sacubitril/ on the clinical stability of patients with heart failure and a
valsartan. Circ Heart Fail 2020;13:e006946. reduced ejection fraction. Circulation 2021;143:326-36.

Jia G, Hill MA, Sowers JR. Diabetic cardiomyopathy: an Pagell RL, O’Bryant CL, Cheng D, et al. Drugs that cause or
update of mechanisms contributing to this clinical entity. exacerbate heart failure: a scientific statement from the
Circ Res 2018;122:624-38. American Heart Association. Circulation 2016;134:e32-69.

Lampert R. Managing with pacemakers and implantable Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin in renal
cardioverter defibrillators. Circulation 2013;128:1576-85. outcomes in type 2 diabetes and nephropathy. N Engl J
Med 2019;380:2295-306.
Lindenfeld J, Zile MR, Desai AS, et al. Haemodynamic-guided
management of heart failure (GUIDE-HF): a randomised Pfeffer MA, Claggett B, Assmann SF, et al. Regional variation
controlled trial. Lancet 2021;398:991-1001. in patients and outcomes in the Treatment of Preserved
Cardiac Function Heart Failure with an Aldosterone Antag-
Maddox TM, Januzzi JL, Allen LA, et al. 2021 Update to the onist (TOPCAT) trial. Circulation 2015;131:34-42.
2017 ACC Expert Consensus Decision Pathway for Opti-
Pitt B, Anker SD, Bushinsky DA, et al. Evaluation of the
mization of Heart Failure Treatment: Answers to 10 Piv-
efficacy and safety of RLY5016, a polymeric potassium
otal Issues About Heart Failure With Reduced Ejection
binder, in a double-blind, placebo-controlled study in
Fraction: A Report of the American College of Cardiol-
patients with chronic heart failure (the PEARL-HF) trial.
ogy Solution Set Oversight Committee. J Am Coll Cardiol
Eur Heart J 2011;32:820-8.
2021;77:772-810.
Pitt B, Filippatos G, Agarwal R, et al. Cardiovascular events
Margulies KB, Hernandez AF, Redfield MM, et al. Effects
with finerenone in kidney disease and type 2 diabetes.
of liraglutide on clinical stability among patients with
N Engl J Med 2021;doi:10.1056/NEJMoa2110956
advanced heart failure and reduced ejection fraction: a
randomized clinical trial. JAMA 2016;316:500-8. Pitt B, Pfeffer MA, Assmann SF, et al. Spironolactone for
heart failure with preserved ejection fraction. N Engl J
Marso SP, Bain SC, Consoli A, et al. Semaglutide and cardio-
Med 2014;370:1383-92.
vascular outcomes in patients with type 2 diabetes. N Engl
J Med 2016b;375:1834-44. Ponikowski P, Kirwan BA, Anker SD, et al. Ferric carboxymalt-
ose for iron deficiency at discharge after acute heart fail-
Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide ure: a multicentre, double-blind, randomised, controlled
and cardiovascular outcomes in type 2 diabetes. N Engl trial. Lancet 2020; 396(10266):1895-1904.
J Med 2016a;375:311-22.
Psotka MA, Gottlieb SS, Francis GS, et al. Cardiac calcit-
McMurray JJV, Packer M. How should we sequence the ropes, myotropes, and mitotropes: JACC review topic of
treatments for heart failure and a reduced ejection frac- the week. J Am Coll Cardiol 2019;73:2345-53.
tion? A redefinition of evidence-based medicine. Circula-
tion 2021;143:875-7. Rich MW, Beckham V, Wittenberg C, et al. A multidisci-
plinary intervention to prevent the readmission of elderly
McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin patients with congestive heart failure. N Engl J Med
in patients with heart failure and reduced ejection frac- 1995;333:1190-5.
tion. N Engl J Med 2019;381:1995-2008.
Riello RJ, Pitt B. ARNI and MRA combination in PARAGON-
Moss AJ, Hall WJ, Cannom DS, et al. Improved survival with HF: odd couple or dynamic duo? JACC Heart Fail 2021;
an implanted defibrillator in patients with coronary dis- 9:25-7.
ease at high risk for ventricular arrhythmia. N Engl J Med
1996;335:1933-40. Shavelle DM, Desai AS, Abraham WT, et al. Lower rates of.
heart failure and all-cause hospitalizations during pulmo-
Moss AJ, Wojciech Z, Hall J, et al. Prophylactic implantation nary artery pressure-guided therapy for ambulatory heart
of a defibrillator in patients with myocardial infarction and failure: one-year outcomes from the CardioMEMS post-­
reduced ejection fraction. N Engl J Med 2002;346:877-83. approval study. Circ Heart Fail 2020;13:229-38.

Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and Solomon SD, McMurray JJV, Anand IS, et al. Angiotensin-­
cardiovascular and renal events in type 2 diabetes. N Engl neprilysin inhibition in heart failure with preserved
J Med 2017;377:644-57. ejection fraction. N Engl J Med 2019;381:1609-20.

PSAP 2022 Book 1 • Cardiology 27 Heart Failure with Reduced Ejection Fraction
Solomon SD, McMurray JJV. Making the case for an Yancy CW, Jessup M, Bozkurt B, et al. 2017 ACC/AHA/HFSA
expanded indication for sacubitril/valsartan in heart focused update of the 2013 ACCF/AHA guideline for the
failure. J Card Fail 2021;27:693-5. management of heart failure: a report of the American Col-
lege of Cardiology/American Heart Association Task Force
Teerlink JR, Diaz R, Felker GM, et al. Cardiac myosin activa- on Clinical Practice Guidelines and the Heart Failure Soci-
tion with omecamtiv mecarbil in systolic heart failure. ety of America. J Am Coll Cardiol 2017;70:776-803.
N Engl J Med 2021;384:105-16.
Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guide-
Vaduganathan M, Fonarow GC, Green SJ, et al. Contempo- line for the management of heart failure: a report of the
rary treatment patterns and clinical outcomes of comor- American College of Cardiology Foundation/American
bid diabetes mellitus and HFrEF: the CHAMP-HF registry. Heart Association Task Force on Practice Guidelines.
JACC Heart Fail 2020;8:459-80. J Am Coll Cardiol 2013;62:e147-e239.
Virani SS, Alonso A, Aparicio HJ, et al; on behalf of the Zannad F, Ferreira JP, Pocock SJ, et al. SGLT2 inhibitors in
American Heart Association Council on Epidemiology and patients with heart failure with reduced ejection fraction:
Prevention Statistics Committee and Stroke Statistics a meta-analysis of the EMPEROR-Reduced and DAPA-HF
Subcommittee. Heart disease and stroke statistics—2021 trials. Lancet 2020;396:819-29.
update: a report from the American Heart Association.
Circulation 2021;143:e254-e743. Zelniker TA, Braunwald E. Mechanisms of cardiorenal effects
of sodium-glucose cotransporter 2 inhibitors: JACC state-
von Haehling S, Ebner N, Evertz R, et al. Iron deficiency in of-the-art review. J Am Coll Cardiol 2020;75:422-34.
heart failure: an overview. JACC Heart Fail 2019;7:36-46.
Zinman B, Wanner C, Lachin JM, et al.; for the EMPA-REG
Wiviott SD, Raz I, Bonaca MP, et al.; for the DECLARE-TIMI 58 OUTCOME Investigators. Empagliflozin, cardiovascular
Investigators. Dapagliflozin and cardiovascular outcomes outcomes, and mortality in type 2 diabetes. N Engl J Med
in type 2 diabetes. N Engl J Med 2018;377:644-57. 2015;373:2117-28.

PSAP 2022 Book 1 • Cardiology 28 Heart Failure with Reduced Ejection Fraction
Self-Assessment Questions
Questions 1–3 pertain to the following case. Updated Universal Definition of HF, which one of the fol-
Q.E., a 57-year-old white woman with a medical history sig- lowing best evaluates Q.E.’s category of HF?
nificant for breast cancer, completed a 6-cycle regimen of
A. HF with reduced ejection fraction (HFrEF)
chemotherapy including doxorubicin, docetaxel, and cyclo-
B. HF with mildly reduced ejection fraction (HFmrEF)
phosphamide less than 1 week ago. She feels fatigued, which
C. HF with improved ejection fraction (HFimpEF)
her oncologist attributes to the recent chemotherapy. Out
D. HF with preserved ejection fraction (HFpEF)
of an abundance of caution, Q.E. is referred to a cardiolo-
gist who performs a laboratory assessment and ECHO that 4. About 4 months ago, a 68-year-old white man was hos-
resulted in an NT-proBNP of 390 pg/mL [reference range <125 pitalized for an anterior wall myocardial infarction com-
pg/mL] and a left ventricular ejection fraction (LVEF) of 58% plicated by left ventricular systolic dysfunction. At
and no identifiable abnormalities. Q.E. denies any symptoms discharge, he was prescribed the following medications:
of heart failure (HF) and reports being able to complete her aspirin 81 mg daily, atorvastatin 80 mg daily, carvedilol
normal activities of daily life without limitations. 12.5 mg twice daily, sacubitril/valsartan 49/51 mg twice
daily, spironolactone 25 mg daily, and ticagrelor 90 mg
1. Which one of the following best evaluates Q.E. with
twice daily. During a post-discharge follow-up visit today,
respect to the Heart Failure Society of America (HFSA),
the patient’s LVEF as measured via ECHO has improved
Heart Failure Association of the European Society of
from 25% to 32% since the last hospital assessment. His
Cardiology (HFA-ESC), and the Japanese Heart Fail-
laboratory results are within normal limits. He is euvole-
ure Society (JHFS) staging system and New York Heart
mic, normotensive, and reports only slight limitations of
Association (NYHA) functional classification?
physical activity. Which one of the following is best to
A. At risk for HF and NYHA Class I recommend for this patient?
B. At risk for HF with no accompanying NYHA
A. Increase carvedilol to 25 mg twice daily.
classification
B. Increase spironolactone to 50 mg daily.
C. Pre-HF and NYHA Class I
C. Start empagliflozin 10 mg daily.
D. Pre-HF with no accompanying NYHA classification
D. Increase sacubitril/valsartan to 97/103 mg twice
2. During her annual follow-up visit to the cardiologist, daily.
Q.E. had a repeat ECHO and laboratory assessment that
5. A 55-year-old white man (weight 214 lb) is admitted to
reported a LVEF of 38% and NT-proBNP of 1670 pg/mL
the hospital with acutely decompensated HF requiring
[reference range <125 pg/mL]. Over the past month, she
inotropic support. Echocardiogram shows LVEF 42%,
reports being unable to walk up a flight of stairs with-
moderate functional mitral regurgitation, and dilated left
out assistance, frequent breaks due to dyspnea while
ventricle. The patient’s medical history is significant for
brushing her teeth or collecting the mail, and requiring
hyperlipidemia and idiopathic dilated cardiomyopathy.
2-3 pillows propped in a chair to sleep a few times per
His condition stabilizes and he is resumed on his home
week. Which one of the following best categorizes Q.E.
regimen of bumetanide 2 mg daily, carvedilol 12.5 mg
according to the HFSA/HFA-ESC/JHFS staging system
twice daily, eplerenone 25 mg daily, lisinopril 20 mg daily,
and NYHA functional classification?
and rosuvastatin 10 mg daily. A basic metabolic panel is
A. HF and NYHA Class II unrevealing aside from a Hgb of 11.2 g/dL. The patient is
B. HF and NYHA Class III clinically stable other than complaints of feeling fatigued
C. Advanced HF and NYHA Class III and inability to participate in physical therapy. He exhib-
D. Advanced HF and NYHA Class IV its no clinical signs of bleeding, so the medical resident
3. Based on Q.E.’s presentation, laboratory, and ECHO find- begins to work up the cause of his anemia. Iron studies
ings, the cardiologist diagnoses her with HF and pre- return with a serum ferritin 115 ng/mL and transferrin sat-
scribes an ARNI and β-blocker that same visit. Q.E. is uration 18% [reference range 20%-50%]. Which one of the
referred to a HF disease management clinic to optimize following is best to recommend to correct this patient’s
the rest of her guideline-directed medical therapy regi- iron-deficiency anemia?
men and titrate to target dosing. After 6 months of consis- A. Ferric carboxymaltose 500 mg IV once
tent follow-up visits in the clinic, a repeat ECHO reports B. Ferrous sulfate 200 mg Monday, Wednesday, Friday
an LVEF of 49%. According to the HFSA/HFA-ESC/JHFS for 3 months
C. Iron dextran 1,000 mg IV once
D. Iron sucrose 200 mg IV every other day for 5 doses

PSAP 2022 Book 1 • Cardiology 29 Heart Failure with Reduced Ejection Fraction
Questions 6 and 7 pertain to the following case. concerns for his renal function. Which one of the follow-
I.M. is a 56-year-old white man with a medical history signif- ing is best to recommend for J.T.?
icant for uncontrolled diabetes and hypertension. He denies
A. Continue without dapagliflozin.
taking any medications and has been lost to follow-up with
B. Initiate empagliflozin at 10 mg daily.
his primary care provider. I.M. presents to the ED with diffi-
C. Initiate empagliflozin at 25 mg daily.
culty breathing due in the setting of volume overload with
D. Reinitiate dapagliflozin at 5 mg daily.
8 kg weight gain in the past week. His laboratory results
are unremarkable except Na: 130 mEq/L, K 4.9 mEq/L, SCr 9. Three months later, J.T. is following-up in the HF disease
2.5 mg/dL (baseline 1.1 mg/dL), and NT-proBNP 11,000 pg/mL management clinic. He is euvolemic with an eGFR 30 mL/
[reference range >300 pg/mL]. Cardiology is consulted and an min/1.73 m2 and reports adherence to all previous med-
ECHO is performed, revealing severely impaired systolic func- ications including an SGLT2 inhibitor. Which one of the
tion with an LVEF of 22%. I.M.’s vital signs are blood pressure following is best to recommend regarding optimization
of 149/88 mm Hg, HR 98 beats/minute, respiratory rate of 24 of J.T.’s guideline-directed medical therapy?
breaths/minute, and oxygen saturation of 97% on room air. A. Increase losartan to 150 mg daily.
I.M. is admitted to the telemetry floor for management. B. Switch losartan to sacubitril/valsartan 49/51 mg
6. The admitting resident is undecided about which medi- twice daily with a 36-hour washout.
cation to start first for I.M.’s acute heart failure manage- C. Switch losartan to sacubitril/valsartan 49/51 mg
ment. Which one of the following is best to recommend twice daily without a 36-hour washout.
for I.M.? D. Increase spironolactone to 50 mg daily.
10. A 47-year-old African American man (weight 176 lb) with
A. Eplerenone
no medical insurance has a medical history significant
B. Furosemide
for drug and alcohol use disorder, hypertension, and
C. Metoprolol succinate
HFrEF (LVEF 31%). He endorses good adherence to his
D. Sacubitril/valsartan
medications and has been stable on a previous regimen
7. After being stabilized, I.M. is initiated on the following of carvedilol 25 mg twice daily, furosemide 20 mg daily,
medication regimen: eplerenone 25 mg daily, furose- lisinopril 40 mg daily, and spironolactone 50 mg daily.
mide 40 mg daily, metoprolol succinate 100 mg daily, On the patient’s last visit to the HF disease management
and sacubitril/valsartan 49/51 mg twice daily. His vital clinic, however, he was switched from lisinopril to sacu-
signs have improved to 124/82 mm Hg, heart rate 62 bitril/valsartan 49/51 mg twice daily. Although he has
beats/minute, respiratory rate 19 breaths/minute, and been tolerating ARNI well, he is about to run out of his
oxygen saturation 99% on room air. A repeat laboratory 30-day free supply provided by a manufacturer coupon
assessment is unrevealing except for a SCr of 1.4 mg/dL. and expresses concerns that he lacks prescription insur-
Which one of the following is best to recommend to opti- ance and cannot afford subsequent refills. The cardiol-
mize I.M.’s guideline-directed medical therapy? ogist transitions him back to lisinopril after a 36-hour
A. Add dapagliflozin 10 mg daily. washout period. Which one of the following is best to
B. Increase eplerenone to 50 mg daily. recommend to optimize this patient’s guideline-directed
C. Increase metoprolol succinate to 150 mg daily. medical therapy?
D. Increase sacubitril/valsartan to 97/103 mg twice A. Reinitiate sacubitril-valsartan at 97/103 mg twice
daily. daily.
B. Initiate dapagliflozin 10 mg daily.
Questions 8 and 9 pertain to the following case. C. Initiate isosorbide dinitrate 20 mg and hydralazine
J.T. is a 72-year-old white man with a medical history signif- 37.5 mg three times daily.
icant for diabetes (A1C 6.8%), CKD stage 4 (baseline SCr 1.3 D. Increase lisinopril to 80 mg daily.
mg/dL) and HFrEF (LVEF 34%). His home drugs include aspi-
11. A 72-year-old white man (weight 168 lb) has a medical
rin 81 mg daily, carvedilol 25 mg twice daily, dapagliflozin 10
history significant for advanced HF (LVEF 21%) with
mg daily, furosemide 20 mg daily, losartan 100 mg daily, met-
LVAD implantation 2 years prior. He is referred to the pal-
formin 1,000 mg twice daily, rosuvastatin 20 mg daily, and spi-
liative care service to discuss goals of care. His home
ronolactone 25 mg daily. J.T. is clinically stable but appears
drugs include bumetanide 4 mg daily, carvedilol 25 mg
overly dry on examination and has had a limited appetite and
twice daily, dapagliflozin 10 mg daily, eplerenone 50 mg
complains of thirst. His laboratory results within normal lim-
daily, ivabradine 7.5 mg twice daily, sacubitril/valsartan
its except for an eGFR 22 mL/min/1.73 m2.
97/103 mg twice daily, and warfarin 6 mg daily. Despite
8. During a visit with J.T.’s primary care provider, dapagli- this regimen, the patient is unable to perform any phys-
flozin and furosemide were both discontinued due to ical activities without significant discomfort and is

PSAP 2022 Book 1 • Cardiology 30 Heart Failure with Reduced Ejection Fraction
predominantly bed bound. He does not wish to undergo activity than normal lately. She can no longer walk the
surgery and wants to “live comfortably” to see his grand- length of the local track without losing her breath and
children graduate high school in 6 months. Which one of sleeps in a chair upright once per week. Which one of the
the following is best to recommend for this patient? following is best to recommend for this patient?
A. Start digoxin 125 mcg daily, goal 0.5-0.9 ng/mL. A. Increase empagliflozin to 25 mg daily.
B. Initiate home infusion of dobutamine 7.5 mcg/kg/ B. Start isosorbide dinitrate 20 mg and hydralazine
min. 37.5 mg three times daily.
C. Initiate home infusion of milrinone 0.125 mcg/kg/min. C. Start spironolactone 25 mg daily.
D. Discontinue GDMT and transition to hospice. D. Start vericiguat 2.5 mg daily.
12. A 57-year-old white man (weight 215 lb) with a medical
history significant for HFpEF (LVEF 55%; NYHA Class III), Questions 14 and 15 pertain to the following case.

well-controlled hypertension, and CKD stage 3 (base- C.H. is a 68-year-old white woman (weight 148 lb) with newly
line SCr 1.2 mg/dL; eGFR 57 mL/min/1.73 m2) arrives diagnosed with HFpEF (LVEF 54%) and a medical history sig-
to the HF diuretic clinic with symptoms of congestion nificant for hypertension and diabetes (A1C 7.4%). Her current
for the first time in the last year. The nurse practitioner medication regimen includes atorvastatin 40 mg daily, chlor-
in the clinic administers intravenous bumetanide 4 mg thalidone 12.5 mg daily, furosemide 20 mg daily as needed,
then performs a medication reconciliation, noting per- and metformin 1000 mg twice daily. C.H.’s blood pressure
tinent home drugs to include amlodipine 10 mg daily, is 128/84 mm Hg and heart rate 65 beats/minute. A labora-
bumetanide 2 mg daily as needed, ferrous sulfate 325 tory assessment for today’s visit includes pertinent results
mg every other day, losartan 50 mg daily, multivitamin of NT-proBNP 750 pg/mL [reference range >125 pg/mL], K 4.5
with B-complex, and rosuvastatin 20 mg daily. Which mEq/L, SCr 1.2, and eGFR 61 mL/min/1.73 m2.
one of the following is best to recommend to optimize 14. Which one of the following is best to recommend to man-
this patient’s HF regimen? age C.H.’s comorbidities?
A. Increase bumetanide 4 mg daily. A. Start empagliflozin 10 mg daily.
B. Start spironolactone 25 mg daily. B. Start liraglutide 0.6 mg subcutaneous daily.
C. Start empagliflozin 10 mg daily. C. Start sitagliptin 25 mg daily.
D. Switch losartan to sacubitril/valsartan 24/26 mg D. Increase chlorthalidone to 25 mg daily.
twice daily.
15. Three months later, C.H. returns for a follow-up appoint-
13. A 61-year-old African American woman (weight 159 lb) ment. Since her last appointment, she has been hos-
was newly diagnosed with HFmrEF (LVEF 44%) after pitalized for acute decompensated HF and required
a hospitalization for myocardial infarction. Her medi- intravenous diuretics. Her vital signs during today’s visit
cal history includes diabetes (A1C = 7.0%). The patient’s are blood pressure 141/88 mm Hg, heart rate 75 beats/
home drugs include aspirin 81 mg daily, clopidogrel 75 minute, and pertinent laboratory results of NT-proBNP is
mg daily, empagliflozin 10 mg daily, furosemide 20 mg 1220 pg/mL [reference range >125 pg/mL], K 4.9 mEq/L,
daily as needed, metformin 1000 mg twice daily, metop- SCr 1.5, and eGFR 29 mL/min/1.73 m2. Which one of the
rolol succinate 100 mg daily, rosuvastatin 20 mg daily, following is best to recommend for C.H.’s HFpEF?
and sacubitril/valsartan 97/103 mg twice daily. Pertinent
A. Start irbesartan 150 mg daily.
laboratory results include K 4.0 mEq/L, SCr 1.1 mg/dL,
B. Start nebivolol 5 mg daily.
and eGFR 65 mL/min/1.73 m2. The patient complains of
C. Start sacubitril/valsartan 24/26 mg twice daily.
increased swelling in her feet and legs with a 4-lb weight
D. Start spironolactone 25 mg daily.
gain over the last few days which she self-medicated with
furosemide. She reports more limitations with physical

PSAP 2022 Book 1 • Cardiology 31 Heart Failure with Reduced Ejection Fraction
Learner Chapter Evaluation: Heart Failure with Reduced Ejection Fraction

As you take the posttest for this chapter, also evaluate the 8. The teaching and learning methods used in the chapter
material’s quality and usefulness, as well as the achievement were effective.
of learning objectives. Rate each item using this 5-point scale: 9. The active learning methods used in the chapter were
effective.
• Strongly agree
10. The learning assessment activities used in the chapter
• Agree
were effective.
• Neutral
• Disagree 11. The chapter was effective overall.
• Strongly disagree 12. The activity met the stated learning objectives.
13. If any objectives were not met, please list them here.
1. The content of the chapter met my educational needs.
2. The content of the chapter satisfied my expectations.
3. The author presented the chapter content effectively.
OTHER COMMENTS
4. The content of the chapter was relevant to my practice 14. Please provide any specific comments related to any
and presented at the appropriate depth and scope. perceptions of bias, promotion, or advertisement of
commercial products.
5. The content of the chapter was objective and balanced.
15. Please expand on any of your above responses, and/or
6. The content of the chapter is free of bias, promotion, and
provide any additional comments regarding this chapter:
advertisement of commercial products.
7. The content of the chapter was useful to me.

PSAP 2022 Book 1 • Cardiology 32 Heart Failure with Reduced Ejection Fraction
Drug-Induced Cardiovascular Disease
By Katherine Aymond, Pharm.D., BCPS, BCCP

Reviewed by Barbara S. Wiggins, Pharm.D., MBA, FCCP, FACC, FNLA, BCPS, BCCP, BCCCP, CLS; and Susan M. Smith, Pharm.D., BCPS

LEARNING OBJECTIVES

1. Develop a treatment plan to monitor for cardiotoxicities associated with cancer therapies.
2. Apply cardio-oncology knowledge to prevent toxicities in patients with cancer.
3. Evaluate an individual patient’s pharmacotherapy to distinguish drug-induced cardiovascular disease.
4. Develop a treatment plan for managing ventricular arrhythmias.
5. Account for potential cardiotoxicities when designing an individualized pharmacotherapy plan.

INTRODUCTION
ABBREVIATIONS IN THIS CHAPTER
The FDA is responsible for regulating drug safety through a series
CNI Calcineurin inhibitor
of pre- and postmarketing surveillance. Over 2 million reports were
CV Cardiovascular
provided in 2020 to the FDA using the Adverse Event Reporting Sys-
CVD CV disease
tem, including drug-induced diseases (fda.gov). The database serves
HF Heart failure
as the FDA’s postmarketing safety surveillance system and is eval-
ICI Immune checkpoint inhibitor
uated by clinical reviewers to identify new safety concerns. These
RAAS Renin-angiotensin-aldosterone
regulations contribute to labeling changes, boxed warnings, precau-
system
tions, and contraindications. Although the U.S. drug approval pro-
SNS Sympathetic nervous system
cess does not allow approval until efficacy and safety are shown in
TdP Torsades de pointes
clinical trials, identification of all possible adverse effects remains a
VEGF Vascular endothelial growth factor
challenge. Rare adverse effects that present as an unrelated condi-
VT Ventricular tachycardia
tion are particularly difficult to recognize. Serious adverse effects,
Table of other common abbreviations. especially those that induce disease, should be readily recognized
by a pharmacist to minimize patient harm. Several classes of medi-
cations have been identified as causing or worsening cardiovascular
disease (CVD). Although these medication classes span a wide array
of pharmacotherapy, agents used in the treatment of oncologic dis-
ease have been implicated in recent years.

CHEMOTHERAPY AND IMMUNOTHERAPY-


INDUCED CARDIOTOXICITY
Medical advances in recent decades in the diagnosis and treatment
of cancer have altered the survivability and morbidity previously
associated with this often-terminal disease. The newly defined field
of cardio-oncology targets CVD as an important cause of death in
many cancer survivors. Many treatment modalities, both old and
new, have cardiotoxicities that can further contribute to the progno-
sis of a patient with cancer. Risk stratification, prevention, identifi-
cation, and treatment of these toxicities can vastly improve patient
care. This timeline spans from cancer prevention strategies and diag-
nosis through survivorship and includes the collaboration of both
cardiology and oncology specialists (Barac 2015).

PSAP 2022 Book 1 • Cardiology 33 Drug-Induced Cardiovascular Disease


Mechanisms and Clinical Manifestations solid tumors. Antiangiogenic targets cause a decrease in nitric
Cardiotoxicity encompasses a wide range of pathophysiologic oxide production, increasing vasoconstriction and peripheral
processes, including arrhythmias, hyper/hypotension, heart vascular resistance and thereby increasing arterial blood pres-
failure (HF), and myocardial ischemia. Possible mechanisms sure (Florescu 2013). Heart failure is one of the most common
for cardiotoxicity include both direct and indirect effects to and earliest-described cardiotoxicities with diagnostic crite-
the CV system. Direct effects include myocardial cell injury by ria. Chemotherapeutic agents are intended to cause apoptosis
the chemotherapeutic or immunotherapeutic agent, resulting and necrosis, suppress growth or angiogenesis, or affect cell
in myocardial dysfunction. Indirect effects encompass a wide repair mechanisms in cancer cells. However, these intended
array of mechanisms including endothelial dysfunction, isch- effects can also damage healthy cardiac cells in the myocar-
emia caused by thrombosis, and hemodynamic alterations. dium, leading to cardiotoxicity. Proposed mechanisms vary by
Molecular pathophysiology is not well understood for some agent, with some direct and indirect postulations, but many
agents and is beyond the scope of this chapter. remain unknown (Yeh 2009).
The effects of cancer-related treatment can range in sever-
ity and persistence, occurring acutely, subacutely, or chron- Agents of Toxicity
ically. Acute and subacute effects often occur between Anthracyclines
therapy initiation and can last up to weeks after therapy
Classified as cytostatic antibiotics, anthracyclines—includ-
completion. These effects may include arrhythmias, acute
ing doxorubicin, daunorubicin, epirubicin, and idarubicin—are
coronary syndromes, pericardial disease, or myocardial dys-
used to treat several solid tumor and hematologic malignan-
function (Florescu 2013). Chronic effects may occur any time
cies (Octavia 2012). Anthracycline-induced cardiomyopathy
after therapy completion and are typically associated with
was first identified in the 1960s and presents as a hypoki-
systolic or diastolic dysfunction leading to HF (Florescu 2013).
netic heart progressing to eventual HF. Several risk factors
A variety of cardiotoxicities exist that can manifest depend-
increase the risk of cardiotoxicity, including age older than 65,
ing on the offending chemotherapeutic or immune-modulating
preexisting heart disease or known CV risk factors, and prior
agent. Arrhythmias, which include both supraventricular (atrial
or concurrent chest irradiation. Risk also increases with the
fibrillation) and ventricular (QT prolongation), occur directly
use of other cardiotoxic chemotherapeutic agents (Cardinale
through chronotropy or indirectly through inflammation.
2020). The mechanisms of toxicity remain unclear for anthra-
Hypertension can coexist in patients with cancer before diag-
cyclines. It has been hypothesized that the cardiomyocyte is
nosis and can be exacerbated by inhibition of angiogenesis, a
the main cellular target within the heart. Possible generation
common target for immunotherapies. Angiogenesis, the devel-
of reactive oxygen species through several mechanisms and
opment of new blood vessels, plays a critical role in growth of
reduction in antioxidant levels induce myocyte damage. This
oxidative stress induces apoptosis of the cardiomyocyte,
leading to cardiomyopathy (Octavia 2012).
BASELINE KNOWLEDGE STATEMENTS Anthracycline-induced cardiomyopathy is classified by
time of onset, including acute, subacute, and chronic. Chronic
Readers of this chapter are presumed to be familiar
with the following: cardiomyopathy, characterized by late-onset HF, is the most
common cardiomyopathy, occurring at a rate of 1.6%–5%.
• General knowledge of various cardiovascular Late-onset HF is defined as occurring at least 1 year after
diseases (CVDs)
treatment completion, up to decades later. Acute toxicity is
• Proposed mechanisms of medications known to
cause CVD rare but is seen immediately after infusion and is reversible.
Findings of both subacute and chronic toxicity include dilated
• General knowledge of management of various CVDs
cardiomyopathy. Anthracyclines are dosed on the basis of
• General knowledge of mechanisms of action of
oncologic agents body surface area, and the risk of HF increases with cumula-
tive dosing. Toxicity is dose-dependent, with higher rates of
Table of common laboratory reference values dysfunction occurring with higher cumulative dose therapy
of doxorubicin greater than 250 mg/m2 or epirubicin greater
ADDITIONAL READINGS than 600 mg/m2 during a lifetime.

The following resource has additional information on Human Epidermal Growth Factor
this topic: Receptor 2 Inhibitors
• Page RL II, O’Bryant CL, Cheng D, et al. Drugs that Human epidermal growth factor 2 (HER2) receptors pro-
may cause or exacerbate heart failure. A scientific mote cell proliferation and are overexpressed in around 25%
statement from the American Heart Association. of breast cancers. Expression of the HER2 receptor causes
Circulation 2016;134:1-38. an aggressive breast cancer associated with a high risk of
recurrence and reduced survival. Human epidermal growth

PSAP 2022 Book 1 • Cardiology 34 Drug-Induced Cardiovascular Disease


factor receptors are also found on the cardiomyocyte. Inhibi- endothelial growth factor inhibitors include bevacizumab for
tion of the HER2 receptor in cardiomyocytes results in abnor- the treatment of colorectal cancers and non–small cell lung
mal growth and repair, leading to contractile dysfunction cancer and sunitinib for metastatic renal cell carcinoma. Tox-
and impaired myocyte survival (Yeh 2009). Trastuzumab is a icity of VEGF inhibitors affects both cardiac and vascular
humanized monoclonal antibody for the HER2 receptor and function and includes hypertension, arterial thromboembo-
is FDA approved for the treatment of HER2-positive breast lism (particularly myocardial infarctions [MIs]), cardiac isch-
cancers. Although trastuzumab-associated cardiotoxicity emia, and cardiac dysfunction. Hypertension incidence is as
can occur with monotherapy, the risk increases dramatically high as 35% in some reported clinical trials (Yeh 2009).
with the addition of an anthracycline-based regimen, regard- The mechanism by which bevacizumab causes hyperten-
less of lifetime cumulative anthracycline dose. Trastuzumab- sion remains unclear. Hypotheses include decreased pro-
associated cardiotoxicity is most associated with a decrease duction of nitric oxide through VGEF inhibition. Nitric oxide
in left ventricular ejection fraction (LVEF) that can progress regulates vascular tone through vasodilation, and abnormal-
to severe HF (Copeland-Halperin 2019). ities in this system remain a target for many CV therapies.
Literature cites the overall incidence of HER2-positive Other hypotheses include alterations in the renin-angio-
breast cancer in up to 25% of patients when trastuzumab is tensin system and cholesterol emboli syndrome. The anti-
used in combination with anthracyclines and as low as 2% angiogenesis effects of VEGF inhibitors can also contrib-
for monotherapy (Yeh 2009). Much like anthracyclines, other ute to endothelial cell dysfunction and cause defects within
risk factors have also been linked to increased risk, includ- the endothelium, activating the clotting cascade. In addition,
ing advanced age, preexisting CVD, and obesity (Copeland- decreased production of nitric oxide can contribute to throm-
Halperin 2019; Yeh 2009). Other anti-HER2 therapies have boembolic events through prostacyclin pathways. Heart
since come onto the market, including lapatinib and pertu- failure associated with bevacizumab is possibly related to
zumab, both of which have increased rates of adverse cardiac uncontrolled hypertension, resulting in progressive contrac-
events, but less often than trastuzumab (Jerusalem 2019; tile dysfunction and eventual cardiac failure (Yeh 2009).
Choi 2017; Swain 2013). A potential risk of cardiotoxicity is
BCR-ABL Kinase Inhibitors
associated with these newer medications; however, experi-
ence is currently limited, and long-term data are needed. Cau- BCR-ABL kinase inhibitors are a subset of tyrosine kinase
tion should be exercised when prescribing anti-HER2 agents inhibitors. These targeted cancer therapies are designed to
to patients with preexisting CVD. inhibit tyrosine kinase signaling pathways, leading to anti-
tumorigenesis and anti-angiogenesis activity. BCR-ABL path-
Vascular Endothelial Growth Factor Inhibitors way activation results in inhibition of apoptosis in chronic
Vascular endothelial growth factor (VGEF) inhibitors target myeloid leukemia, allowing chronic myeloid leukemia cells to
angiogenesis, which is essential for tumor growth. Vascular evade the body’s natural response (Chen 2008). All approved

Patient Care Scenario


A 45-year-old woman with a medical history of refractory HF, pulmonary embolism, and chronic obstructive pulmo-
multiple myeloma, hypertension, and smoking presents at nary disease exacerbation. During your discussion with
the clinic with new-onset shortness of breath, dyspnea on the primary physician, the team asks whether any medi-
exertion, lower extremity pitting edema, and inability to cations could be causing the patient’s symptoms. What
complete household tasks over the past several weeks. The strategy would be best to recommend for this patient?
team begins a workup for potential diagnoses, including

ANSWER
It is important to perform a thorough medication his- drug-induced CVD is suspected, the pharmacist should
tory to determine which medications she has previously investigate the temporal association between the intro-
been treated with. The pharmacist should specifically ask duction of the drug in question and symptom onset,
about chemotherapy, immunotherapy, preventive treat- factoring in baseline information, if available. In addi-
ments, and radiation that may have been used to treat tion, other causative factors should be excluded.
her cancer. This information will help identify whether Discontinuation of the drug in question may be recom-
any of these treatments are a likely cause of the drug-in- mended if a specific drug-induced CVD is suspected and
duced CVD contributing to her current condition, such as alternative therapy can be used. Referral to a cardio-on-
anthracycline-induced cardiotoxicity, VEGF inhibitor car- cologist should be provided.
diotoxicity, or corticosteroid-induced hypertension. If

1. Alexandre J, Cautela J, Ederhy S, et al. Cardiovascular toxicity related to cancer treatment: a pragmatic approach to the American and
European cardio-oncology guidelines. J Am Heart Assoc 2020;9:e018403.

PSAP 2022 Book 1 • Cardiology 35 Drug-Induced Cardiovascular Disease


agents are active against BCR-ABL, but with unpredictable toxicity is myocarditis, occurring at a rate of 1%–2% with an
levels of potency and activity for other kinases (Aghel 2017). associated mortality of 27%–46% (Michel 2019). Conduc-
Agents in this class—including imatinib, dasatinib, nilotinib, tion abnormalities from myocarditis include complete heart
bosutinib, and ponatinib—provide “on-target” toxicity to the block, supraventricular tachycardias, and ventricular tachy-
intended cancer cells contributing to apoptosis and anti- cardias (VTs), with VTs leading to sudden death (Brahmer
proliferation pathways. “Off-target” toxicities also occur at 2018). Inflammation can also lead to progressive loss of left
unintended cell lines with kinase pathways, such as the car- ventricular function, resulting in HF. Development of myo-
diomyocyte, resulting in dysfunction (Chen 2008). The vari- carditis is typically observed in the subacute phase within
able potency and activity therefore lead to different safety 1 month of initiation. The exact mechanism remains unde-
profiles for each agent. The detailed mechanism for each fined. Other cardiotoxicities noted in the literature include
agent is beyond the scope of this chapter. Takotsubo syndrome, pericarditis, and acute coronary syn-
Cardiotoxicity for the BCR-ABL kinase inhibitor class of dromes. Both Takotsubo syndrome and pericarditis are sec-
drugs encompasses several different pathologies, including, ondary pathologies associated with several kinds of cancers;
but not limited to, edema, hypertension, hypotension, arrhyth- therefore, evaluation of these patients should include careful
mias, QTc prolongation, pericardial effusions, and left ventric- elimination of other causes. The pathogenesis of acute cor-
ular dysfunction. The most cardiotoxic agent in this class is onary syndrome in patients using ICIs is hypothesized to be
the third-generation agent ponatinib. Ponatinib is responsi- mediated by existing plaque disruption (Michel 2019).
ble for both adverse vascular effects and cardiotoxicities.
Ponatinib’s lack of selectivity for cancer cell kinases has led Ibrutinib
to significant cardiotoxicity through several pathways in the Ibrutinib is a first-in-class agent inhibiting Bruton tyrosine
cardiomyocyte. Ponatinib’s inhibition results in impaired sur- kinase, which is responsible for the growth and proliferation
vival of the cardiomyocyte, resulting in loss of cells and car- of B-cell cancers. Ibrutinib is an effective first-line treatment
diac dysfunction (Singh 2020). The vascular effects were for chronic lymphocytic leukemia. Although ibrutinib was pre-
noted in preclinical trials, with an excess of events noted in a viously considered well tolerated, recent meta-analyses have
long-term follow-up trial. Several serious occlusive events— shown an increased risk of atrial fibrillation at a rate of 3.3 up
including coronary artery occlusions, MIs, cerebrovascu- to 5.77 per 100 person-years (More 2020; Ganatra 2018). In
lar occlusions, peripheral arterial occlusions, and venous addition, other cardiotoxicities can occur, including ventricu-
thromboembolisms – led to suspension of ponatinib’s FDA lar arrhythmias and hypertension.
approval in October 2013. However, the suspension was lifted The mechanism of pathogenesis is not well understood. It
with added monitoring, and a boxed warning was added to the is postulated that Bruton tyrosine kinase is expressed in car-
prescribing information in December 2013 because ponatinib diac tissue, particularly in the atria, and may play a role during
is the only treatment for patients with chronic myeloid leuke- stress conditions (Ganatra 2018).
mia who have a T3151 mutation (Singh 2020). Ibrutinib-associated atrial fibrillation should be man-
aged in accordance with the most recent American College
Immune Checkpoint Inhibitors of Cardiology (ACC) guidelines. Rate control with β-blockers
Immune checkpoint inhibitors (ICIs) encompass several dif- is preferred as initial treatment. Calcium channel blockers,
ferent drugs that have transformed the management of var- verapamil, and diltiazem should be avoided because of drug-
ious cancers. Checkpoint pathways are innate mechanisms drug interactions. Treatments for rhythm control can be dis-
within the human body to control immune responses. Two pro- cussed with the patient if rate control is inadequate or the
teins are considered checkpoint proteins – cytotoxic T-lym- patient is hemodynamically unstable, and no drug-drug inter-
phocyte-associated protein 4 and programmed cell death actions exist (Alexandre 2020).
protein 1 – and are found on the surface of cytotoxic T cells. Ibrutinib-associated atrial fibrillation together with cancer-­
Immune checkpoint inhibitors prevent binding of receptors associated thromboembolism poses a complex challenge for
and ligands and inhibit signaling pathways used by cancer clinicians to balance the efficacy and safety of anticoagula-
cells to evade T cell–mediated death in normal immune func- tion. New-onset atrial fibrillation associated with malignancy
tion. Six ICIs are FDA approved for the treatment of advanced has an increased risk of thromboembolism and HF. Cancer
malignant cancers, including first-to-market agents ipilim- is a well-defined thromboembolic risk factor, and clinical tri-
umab, nivolumab, and pembrolizumab for the treatment of als have often excluded patients with active malignancy.
unresectable or metastatic melanoma. In a large retrospective cohort, new-onset atrial fibrillation
Several toxicities are associated with ICIs and are called after a cancer diagnosis was associated with higher rates
immune-related adverse effects. Modulation of immune func- of HF and thromboembolism (Hu 2013). In patients with can-
tion through T-cell activity affects several organ systems in cer with new-onset atrial fibrillation, an increasing CHADS2
the body and can range from mild, allowing for treatment score (greater than 4) was associated with higher mortality
continuation, to severe, resulting in death. The most lethal but not increased rates of thromboembolism (Hu 2013). In

PSAP 2022 Book 1 • Cardiology 36 Drug-Induced Cardiovascular Disease


Patient Care Scenario
A 45-year-old woman (weight 80 kg) has a medical his- A. Discontinue ibrutinib and refer her to a cardiologist.
tory that includes hypertension and chronic lymphocytic B. Administer intravenous diltiazem 10 mg once and
leukemia. She presents at the ED in atrial fibrillation. She initiate a systemic heparin infusion.
has concerns about dizziness, with heart rate 130 beats/ C. Administer intravenous metoprolol 5 mg once.
minute and blood pressure 130/84 mm Hg. Her SCr is 1.2
D. Administer intravenous amiodarone 150 mg once
mg/dL. Her current medications include ibrutinib 420 mg
followed by continuous infusion and apixaban 5 mg
once daily and lisinopril 10 mg daily. Which one of the
twice daily.
following is best to recommend to manage this patient’s
atrial fibrillation?

ANSWER
Rate control with β-blockers is preferred in patients guideline recommendations. When ibrutinib is one of the
with atrial fibrillation on ibrutinib (Answer C is correct). only options for cancer, therapy should be continued and
Rate control with non-dihydropyridine calcium chan- referral for evaluation should be made to a cardio-oncol-
nel blockers is contraindicated because diltiazem is a ogist. Initiating rhythm control would be inappropriate in
CYP 3A4 inhibitor and will increase toxicity of ibrutinib. this patient as rate control is preferred first line for hemo-
In addition, this patient’s CHA 2DS2-VASc score is 2 and dynamically stable patients (Answer A, Answer B, and
does not qualify her for anticoagulation under current Answer D are incorrect).

1. Imbruvica (ibrutinib). Package insert. Janssen Biotech, July 2019.


2. Alexandre J, Cautela J, Ederhy S, et al. Cardiovascular Toxicity Related to Cancer Treatment: A Pragmatic Approach to the American and
European Cardio-Oncology Guidelines. J Am Heart Assoc 2020;9:e018403.
3. January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management
of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical
Practice Guidelines and the Heart Rhythm Society in Collaboration With the Society of Thoracic Surgeons [published correction appears
in Circulation. 2019 Aug 6;140(6):e285]. Circulation. 2019;140(2):e125-e151.

addition, newer stroke risk estimators such as CHA 2DS2VASc identification, with risk factors including chemotherapy expo-
have been shown to underestimate the risk of thromboembo- sure and its related toxicities. Developed in 2016, the Ameri-
lism (D’Souza 2018). This highlights that stroke risk assess- can Society of Clinical Oncology (ASCO) guidelines address
ment tools should be interpreted with caution in patients with cardiac dysfunction in adult cancer survivors. The advisory
cancer and that decisions to use anticoagulation should be group sought to answer five questions that address (1) risk
individualized. categorization, (2) prevention before initiation of chemo-
Anticoagulation selection for ibrutinib-associated atrial therapy, (3) prevention during chemotherapy, (4) monitoring
fibrillation is challenging because of the many drug-drug during chemotherapy, and (5) monitoring after chemotherapy
interactions with commonly used medications. Ibrutinib is (Armenian 2016). Risk categorization should be addressed in
metabolized by CYP3A4 and CYP2D6. According to the pack- all patients regardless of their treatment phase.
age insert, ibrutinib is a weak inducer of CYP3A4 and inhibits The guideline authors chose to focus on the develop-
P-glycoprotein (P-gp) (Imbruvica 2019). Increased bleeding ment of systolic rather than diastolic impairment because of
risk has been identified with the use of ibrutinib because of the lack of data available to distinguish management in the
effects on platelet signaling pathways and thrombocytope- respective patient populations. Systolic cardiac impairment
nia. Choice of anticoagulant remains controversial. Oral Xa presents as a decrease in the LVEF and can be symptomatic
inhibitors have clinically insignificant CYP3A4 interactions or asymptomatic. Other cardiotoxicities such as coronary
with ibrutinib. Warfarin is also an option; however, it has artery disease or pericardial and valvular pathophysiology
been shown to increase major hemorrhage. Warfarin remains are not discussed.
desirable because of its monitoring ability to ensure adequate In 2018, ASCO in partnership with the National Comprehen-
anticoagulation. Dabigatran is a major substrate for P-gp and sive Cancer Network published an additional practice guide-
is contraindicated. Careful consideration for thromboem- line focused specifically on immune-related adverse effects
bolic risk and bleeding should be weighed for each patient, related to patients using ICIs. This guideline discusses man-
allowing for shared decision-making (Ganatra 2018). agement of all immune-related adverse effects associated with
ICI therapy in all major organ systems, including CV. The guide-
American Society of Clinical Oncology line aims to answer the broad question, “How should clinicians
Guidelines manage immune-related adverse effects in adult patients with
The ACC and American Heart Association (AHA) describe cancer treated with immune checkpoint blockade antibodies?”
HF as a progressive disease that starts with risk factor (Brahmer 2018). Recommendations are not graded as usually

PSAP 2022 Book 1 • Cardiology 37 Drug-Induced Cardiovascular Disease


found in the guidelines; rather, recommendations represent a
Box 1. Risk Factors for CVD
consensus opinion, with dissent noted in the text.
The guideline recognizes the most common CV toxicities Modifiable Risk Factors
Diabetes
as myocarditis, pericarditis, arrhythmias, impaired ventricu-
Dietary intake
lar function with HF, and vasculitis. The guideline also details Dyslipidemia
recommendations for venous thromboembolism manage- Hypertension
ment. The authors note that although CV toxicities are not the Physical inactivity
most common toxicity associated with ICIs, they are associ- Premature CVD in a first-degree relative:
ated with significant morbidity and mortality. Cardiotoxicities • < 55 yr in male relative
are noted for all agents within the class, but data are limited • < 65 yr in female relative
to case reports and small case series, encompassing less Tobacco use
than 0.1% of patients (Brahmer 2018). Nonmodifiable Risk Factors
Age:
Prevention
• Males ≥ 45 yr
Risk of cardiac dysfunction includes both treatment- • Females ≥ 55 yr
specific risk factors and non–treatment-related modifiers Sex
of risk. Patients treated with high-dose anthracyclines are • Male
at increased risk of developing cardiotoxicity. Lower doses • Post-menopausal women
of anthracyclines combined with trastuzumab in addition to
CVD = cardiovascular disease.
CV risk factors also increase a patient’s risk of cardiac dys-
Information from: Arnett DK, Blumenthal RS, Albert MA,
function. Older age contributes to this increased risk, with et al. 2019 ACC/AHA guideline on the primary preven-
patients 65 and older implicated most often. Common CV risk tion of cardiovascular disease: a report of the American
factors and comorbidities contribute to the overall cardiac College of Cardiology/American Heart Association
health of the patient with cancer, and the presence of such Task Force on Clinical Practice Guidelines. Circulation
risk factors can contribute to cardiotoxicities. Two or more 2019;140:e596-e646.
modifiable risk factors such as obesity, hypertension, diabe-
tes, dyslipidemia, and smoking increase the risk of HF. Pres-
selection of agents, type and stage of cancer, and efficacy of
ence of compromised heart function at baseline substantially
other available treatments.
increases the risk of further dysfunction. Patients with bor-
There are no strategies to prevent the development of car-
derline low ejection fractions (EFs), a history of MI, or mod-
diotoxicity before initiation of these specific agents. Baseline
erate valvular heart disease are all considered compromised.
monitoring helps identify ineligible candidates for therapy
These risk modifiers should be considered before initiating
and serves as a comparator during treatment. Consensus
specific cardiotoxic therapies such as anthracyclines or tras-
and expert opinion suggest avoidance of therapies with the
tuzumab (Armenian 2016).
potential to cause cardiotoxicity if an alternative treatment is
Before initiation of potential cardiotoxic therapies, the
available. Comprehensive assessment of the patient before
AHA recommends a cardio-oncologic evaluation for patients
initiation is also recommended (Armenian 2016).
using anthracyclines, HER2 inhibitors, VEGF inhibitors, ibru-
Risk of cardiotoxicity during drug administration can be
tinib, and certain BCR-ABL inhibitors. Patients using ponati-
minimized by actively managing modifiable risk factors. Anth-
nib and nilotinib should be referred for evaluation, whereas
racycline-induced cardiomyopathy can be managed using
users of imatinib, bosutinib, and dasatinib need be referred
several strategies. Dexrazoxane interferes with free radical
only if they are at very high CV risk (Box 1) (Alexandre 2020).
generation and is cardioprotective against anthracycline-in-
Cardio-oncologic evaluation is only necessary in patients
duced cardiotoxicity. Dexrazoxane can be administered
using ICIs if they have cardiotoxic effects after initiation. The
before anthracycline infusions and has shown significant risk
AHA recommends that a cardio-oncologic evaluation include
reduction for acute HF in patients receiving high-dose anth-
CV imaging and testing together with a physician visit. This
racyclines (Cardinale 2020). Benefit in lower-dose anthracy-
cardio-oncologic evaluation includes ECG, lipid panel, trans-
cline regimens has not been determined; therefore, they are
thoracic echocardiogram including LVEF measurements,
not commonly used in clinical practice (Armenian 2016). Dex-
active management of modifiable cardiac risk factors, and
razoxane should be considered to prevent cardiotoxicity in
encouragement of a heart-healthy lifestyle, including diet
patients who have advanced metastatic breast cancers with
and exercise counseling. Baseline cardiac biomarkers such
cumulative anthracycline doses greater than 250 mg/m2 of
as high-sensitivity troponin I, BNP, and N-terminal pro–brain
doxorubicin and greater than 600 mg/m2 of epirubicin (Arme-
natriuretic peptide (NT-proBNP) may also be useful before
nian 2016). Using continuous infusion dosing rather than bolus
initiating therapy (Koutsoukis 2018). If baseline abnormali-
dosing of anthracyclines has also decreased cardiotoxicity.
ties are detected, careful consideration should be placed on

PSAP 2022 Book 1 • Cardiology 38 Drug-Induced Cardiovascular Disease


Liposomal formulations of doxorubicin are also associated short-term follow-ups. Since publication of the 2016 ASCO
with decreased risk. These three strategies show benefit in the guidelines, several randomized control trials and meta-
setting of high cumulative dosing of anthracyclines but are of analyses have been conducted and published; however,
unknown benefit in patients receiving lower dosing regimens. results continue to show mixed benefit (Table 1).
Use of angiotensin-converting enzyme (ACE) inhibitors, Results of the CECCY trial, published in 2018, showed no
β-blockers, and angiotensin receptor blockers (ARBs) as pro- difference in the rate of LVEF decrease among patients with
phylaxis for cardiomyopathy has been explored in recent HER2-negative breast cancer prophylactically treated with
years. One early study showed that early initiation of enal- carvedilol compared with placebo (14.5% vs. 13.5%, p=1.0)
april in patients with elevated troponin (“troponin triggered”) (Avila 2018). After publication of CECCY, a meta-analysis
receiving high-dose anthracyclines prevented a decrease concluded that β-blockers preserved LVEF by almost 4%.
in LVEF (Cardinale 2006). Most trials evaluating the effects The population was mixed but mainly included patients with
of standard HF therapies as prevention for chemotherapy- breast cancer and excluded the use of ACE inhibitors and
induced toxicities have included small sample sizes with trastuzumab (Shah 2019).

Table 1. Summary of Trials Evaluating ACE Inhibitor, ARB, or β-Blocker Prophylaxis for Anthracycline-Induced Cardiotoxicity
Since 2016 ASCO Guideline Publication

Study/Design Treatment Outcome Results Conclusion

Avila 2018 Carvedilol vs. placebo Prevention of ≥ 10% LVEF 14.5% vs. 13.5% (p=1.0) No benefit
CECCY trial reduction

Shah 2019 β-Blocker vs. placebo Final LVEF -3.84 mean difference β-Blocker associated
Meta-analysis with preservation of EF
by almost 4%

Cardinale 2018 Enalapril prevention Troponin elevation above 23% vs. 26% (p=0.50) No benefit between
ISCO-1 trial vs. troponin-triggered threshold strategies
enalapril

Georgakopoulos Metoprolol vs. Occurrence of clinical or No clinical symptoms Coadministration of


2019 enalapril vs. placebo subclinical cardiotoxicity of HF metoprolol or enalapril
offers no benefit

Słowik 2020 Ramipril vs. placebo Occurrence of cardiotoxicity 6.3% vs. 18.5% (p=0.15) No benefit on LVEF
defined by biomarkers or
decrease 10% of LVEF

Lee 2021 Candesartan vs. Decrease in LVEF > 10% 4.9% vs. 8.6% vs. 18.6% Candesartan may be
carvedilol vs. placebo within 6 mo of therapy (p=0.022 for candesartan effective in preventing
vs. placebo, p=0.145 for early decrease in LVEF
carvedilol vs. placebo)

Lin 2021 ACE inhibitors/ARBs Change in LVEF -3.16 mean difference ACE inhibitors and ARBs
Meta-analysis vs. controls reduced cardiotoxicity
by preserving LVEF

CECCY = Carvedilol Effect in Preventing Chemotherapy-Induced Cardiotoxicity; ISCO = International CardioOncology Society-One
(trial); LVEF = left ventricular ejection fraction.
Information from: Avila MS, Ayub-Ferreira SM, de Barros Wanderley MR Jr, et al. Carvedilol for Prevention of Chemotherapy-Related
Cardiotoxicity: The CECCY Trial. J Am Coll Cardiol. 2018;71(20):2281-2290, Shah P, Garris R, Abboud R, et al. Meta-Analysis Comparing
Usefulness of Beta Blockers to Preserve Left Ventricular Function During Anthracycline Therapy. Am J Cardiol. 2019;124(5):789-794,
Cardinale D, Ciceri F, Latini R, et al. Anthracycline-induced cardiotoxicity: A multicenter randomised trial comparing two strategies for
guiding prevention with enalapril: The International CardioOncology Society-one trial. Eur J Cancer. 2018;94:126-137, Georgakopoulos
P, Kyriakidis M, Perpinia A, et al. The role of metoprolol and enalapril in the prevention of doxorubicin-induced cardiotoxicity in
lymphoma patients. Anticancer Res 2019;39:5703-7; Słowik A, Jagielski P, Potocki P, et al. Anthracycline-induced cardiotoxicity
prevention with angiotensin-converting enzyme inhibitor ramipril in women with low-risk breast cancer: results of a prospective
randomized study. Kardiol Pol 2020;78:131-7; Lee M, Chung WB, Lee JE, et al. Candesartan and carvedilol for primary prevention of
subclinical cardiotoxicity in breast cancer patients without a cardiovascular risk treated with doxorubicin. Cancer Med 2021;10:
3964-73; Lin H, Liang G, Wu Y, et al. Protective effects of ACEI/ARB on left ventricular function in anthracycline-induced chronic
cardiotoxicity: a meta-analysis of randomized controlled trials. Cardiology 2021;146:469-80.

PSAP 2022 Book 1 • Cardiology 39 Drug-Induced Cardiovascular Disease


The role of renin-angiotensin system inhibition has been Current guideline recommendations do not include the use
explored in several trials. The ISCO-1 trial conducted in Italy of prophylactic ACE inhibitors, ARBs, or β-blockers for anth-
randomly assigned patients to receive enalapril prophylacti- racycline- or trastuzumab-treated patients because of lack of
cally (“prevention” arm) or after an increase in troponin (tropo- consistent evidence and need for larger and longer clinical tri-
nin-triggered arm). The primary outcome of troponin elevation als. However, as evidenced by meta-analyses, early β-blocker
occurred in 23% in the prevention groups and 26% in the tro- treatment potentially has short-term beneficial effects in pre-
ponin-triggered group (p=0.50), showing that ACE inhibitor serving LVEF and is reasonable to consider (Blanter 2019).
treatment may only be necessary in patients with troponin Prophylactic use of ACE inhibitors or ARBs has shown mod-
elevation rather than prophylactic use (Cardinale 2018). est changes in LVEF, but further, large-scale studies are
Investigators sought to define the long-term benefits of needed (Lin 2021).
prophylactic therapy with metoprolol or enalapril in patients
with lymphoma treated with anthracycline-based regimens. Diagnosis and Treatment
Patients were randomly assigned to placebo, metoprolol, or
Initial Evaluation
enalapril open label and followed for 10 years. The primary
Neither the frequency of monitoring nor its effect on out-
outcome of clinical or subclinical cardiotoxicity did not differ
comes is defined for patients at risk of cardiotoxicity.
between the three groups at the 3- or 10-year mark. Like previ-
Frequent symptom monitoring in all patients exposed to car-
ous trials, improvements in diastolic dysfunction were noted
diotoxic anticancer agents should occur throughout treat-
in the metoprolol group (Georgakopoulos 2019).
ment and follow-up. Patients who have previously received
Investigators prospectively studied treatment with ramipril
anthracycline therapy should be considered for echocardi-
shortly before chemotherapy in young women with no CV risk
ography within 1 year of therapy completion. Patients with
factors newly diagnosed with breast cancer. They randomly
evidence of structural heart disease on echocardiography,
assigned patients to ramipril or placebo and estimated the
including left ventricular dysfunction, hypertrophy, valvular
LVEF at baseline. The primary outcome was a combination
disease, or left ventricular dilation, regardless of symptoms,
of increased concentrations of troponin or NT-proBNP and a
should be staged using classifications set forth by the ACC
decrease in LVEF of more than 10%. There were no significant
and AHA guidelines for HF management. Referral to a cardiol-
differences between the groups. The authors concluded that
ogist should occur for stages B, C, and D (NCCN 2021).
relatively healthy young women with breast cancer would
Anthracycline-induced cardiotoxicity is defined by a
likely not benefit from ACE inhibitor therapy (Słowik 2020).
decrease in LVEF of at least 10% from baseline with an overall
Other investigators sought to determine whether ARB or
decrease of less than 50%. In cardio-oncology, toxicities are
β-blocker therapy would mitigate the risk of left ventricular
further classified as type I or type II. Type I is considered irre-
dysfunction early in the treatment of healthy patients with
versible, as seen with anthracycline use. Type II is associated
breast cancer taking anthracyclines. This was a post hoc
with targeted therapies and is considered reversible.
analysis of the SAVE HEART trial to evaluate the effects of
candesartan or carvedilol compared with a control group, Imaging and Monitoring
which was omitted from the initial analysis. The primary Patients noted to be at high risk of CVD should undergo more
outcome was early doxorubicin-induced subclinical cardio- frequent monitoring while receiving cardiotoxic chemother-
toxicity (DISC) defined by development within 6 months of apeutic agents. Routine surveillance for all patients should
chemotherapy. Doxorubicin-induced subclinical cardiotoxic- include a careful history and a complete physical examina-
ity was defined by an LVEF decrease of greater than 10% or tion at each visit. Referral to a cardio-oncologist is also rec-
below 50% at follow-up. Candesartan significantly reduced ommended for further evaluation. Alternative treatments
the incidence of early DISC compared with control (4.9% vs. should be considered for patients experiencing symptoms
18.6%, p=0.022). The authors concluded that low-dose can- at any time during therapy while weighing the risk-benefit
desartan had a lower incidence of early DISC in patients with (Domercant 2016). Echocardiography should be performed
breast cancer with no CV risk (Lee 2021). for patients experiencing symptoms to rule in or out HF symp-
Investigators conducted a meta-analysis of randomized tomology. Electrocardiograms should be obtained in patients
controlled trials using ACE inhibitors and ARBs prophylacti- experiencing arrhythmias.
cally to prevent anthracycline-induced cardiotoxicity. They
included seven trials using change in LVEF as their end Follow-up and Treatment
point. They found that use of prophylactic renin-angiotensin- Interruption of chemotherapy for toxicities may be necessary,
aldosterone system (RAAS) inhibition better preserved LVEF, depending on the severity. Treatment for each specific toxic-
with a mean difference of -3.16% (p=0.02). They concluded ity does not differ from that for patients without cancer. For
that although there were only a few clinical trials and partic- example, patients found to have a decrease in LVEF with signs
ipants, use of ACE inhibitors or ARBs reduced cardiotoxicity and symptoms of HF should be treated in accordance with
associated with anthracyclines (Lin 2021). the ACC and AHA guidelines. Clinicians must also carefully

PSAP 2022 Book 1 • Cardiology 40 Drug-Induced Cardiovascular Disease


Table 2. Summary of Specific Chemotherapeutic Agents and Prevention or Mitigation Strategies for Cardiotoxicities

Associated
Agent/Class Cardiotoxicity Prevention or Mitigation Strategies Other Pearls

Anthracyclines Cardiomyopathy All dosing: Do not initiate for LVEF < 40%
HF • Cardio-oncologic evaluation at baseline, end of unless no other effective
treatment, 6 mo, 1 yr, and 2 yr treatment available
• Baseline BNP, troponin
• Optimal management of CV risk factors
High dose:
• Consider dexrazoxane
• Additional cardio-oncologic evaluation before
each cycle
• Troponin at the end of each cycle
Low dose:
• Troponin at the end of each cycle if risk factors
present
CV symptoms or positive troponin:
• Cardio-oncologic evaluation

HER2 inhibitors Cardiomyopathy • Cardio-oncologic evaluation at baseline, every Do not initiate for LVEF < 40%
Trastuzumab HF 3 mo during treatment, end of treatment, 6 mo, unless no other effective
1 yr and 2 yr treatment available
• Baseline BNP, troponin
• Optimal management of CV risk factors
• Troponin after each cycle if risk factors present
CV symptoms or positive troponin:
• Cardio-oncologic evaluation

VEGF inhibitors Hypertension • Cardio-oncologic evaluation at baseline, every


Myocardial ischemia 3 mo for 1 yr, every 6 mo during treatment
LV dysfunction • Optimal management of CV risk factors
QTc prolongation • Home BP monitoring
Arterial thromboem-
bolic events

BCR-ABL kinase Atherosclerosis • Cardio-oncologic evaluation at baseline, every Nilotinib carries a boxed
inhibitors PAD 3 mo for 1 yr, and every 6 mo during treatment warning for QT prolongation
ACS for ponatinib and nilotinib and high-risk patients and sudden death. Ponatinib
Stroke taking imatinib, bosutinib, and dasatinib has been associated with
Pericardial effusion • Optimal management of CV risk factors severe arterial thrombotic
Pulmonary artery • Home BP monitoring for ponatinib and nilotinib events during treatment.
hypertension • PAD screening and ankle-brachial index with
QTc prolongation Doppler of lower extremities at baseline for
nilotinib and every 6 mo if at high risk, for both
ponatinib and nilotinib

ICIs Immune-related • ECG and troponin at baseline, within 48 hr of Grading (G) levels 1–4
myocarditis each administration, and if symptomatic G1: Abnormal testing
Pericarditis • Cardio-oncologic evaluation if positive troponin (biomarkers or ECG)
Supraventricular or ECG abnormal G2: G1 + mild symptoms
arrhythmias G3: G1 + symptoms with mild
ACS activity
Takotsubo G4: Moderate to severe
syndrome decompensation, life
threatening
Permanently discontinue ICIs
for all grades of cardiotoxicity

(continued)

PSAP 2022 Book 1 • Cardiology 41 Drug-Induced Cardiovascular Disease


Table 2. Summary of Specific Chemotherapeutic Agents and Prevention or Mitigation Strategies for Cardiotoxicities
(continued)

Ibrutinib AF • Cardio-oncologic evaluation at baseline, every AF most likely in the first


Ventricular 3 mo for 1 yr, and every 6 mo during treatment 30 days
arrhythmias • Home BP monitoring
HF • Symptomatic patients monitored with Holter
Hypertension ECG

ACS = acute coronary syndromes; AF = atrial fibrillation; BP = blood pressure; HF = heart failure; ICI = immune checkpoint inhibitor;
LV = left ventricular; PAD = peripheral arterial disease; VEGF = vascular endothelial growth factor.
Information from: Alexandre J, Cautela J, Ederhy S, et al. Cardiovascular toxicity related to cancer treatment: a pragmatic approach
to the American and European cardio-oncology guidelines. J Am Heart Assoc 2020;9:e018403; Brahmer JR, Lacchetti C, Schneider
BJ, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American
Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2018;36:1714-68.

weigh other patient-specific factors such as drug-drug inter-


actions, laboratory assessments (for hematologic abnormal- Table 3. Medications Associated with Drug-Induced
ities), and patient outcomes. Table 2 summarizes prevention Sinus Bradycardia and AV Nodal Blockade
and mitigation strategies.
Sinus AV Nodal
Medication Bradycardia Blockade
OTHER AGENTS OF CARDIOTOXICITY Adenosine X X
Arrhythmias
Amiodarone X X
Supraventricular Arrhythmias β-Blockers X X
Sinus Bradycardia and Atrioventricular Nodal
Clonidine X
Blockade
Dexmedetomidine X
Sinus bradycardia is defined as a heart rate less than 60
beats/minute. Dysfunction of the sinoatrial (SA) node, which Digoxin X X
is responsible for the automaticity of the heart, can also Dronedarone X
result in sinus “pauses” or sinus arrest. Atrioventricular (AV)
Ivabradine X X
nodal blockage occurs when electrical impulses do not prop-
agate from the atria and ventricles. Blockade is described Non-DHP CCB X X
as first, second, or third degree on the basis of severity and Propafenone X X
characteristics. First-degree AV block is a delay in conduc-
tion resulting in a lengthened PR interval by 0.2 seconds. AV = atrioventricular; non-DHP CCB = non-dihydropyridine
Second-degree AV block is exhibited by a lack of conduction calcium channel blocker.

of some impulses between the atria and the ventricle, result- Information from: Wiggins BS, Lehner AL. Drug-induced
cardiovascular disease. In: Jackevicius C, Patterson JH,
ing in a “dropped” beat. Third-degree AV block, or complete eds. Cardiology Self-Assessment Program, 2020 Book 1.
heart block, is exhibited by no association between atria and Critical Care Cardiology. American College of Clinical
ventricular beats. Pharmacy, 2020:93-119.
Several mechanisms can cause drug-induced sinus bra-
dycardia, including SA node automaticity inhibition, slow SA
node conduction, or prolonged repolarization of the SA node. accumulation because of decreased elimination or metab-
Atrioventricular block can be caused by AV node conduction olism, and synergistic or additive effects of combining drug
inhibition or prolonged repolarization of the AV node. In addi- therapies.
tion, activation or inhibition of sympathetic and parasympa- Prevention of sinus bradycardia and AV nodal blockage
thetic nervous systems can affect conduction. Drugs that is imperative. Careful monitoring of heart rate is required in
affect the action potentials – including calcium channel and those with first-degree heart block but is not a contraindica-
sodium channel inhibitors – may also cause sinus bradycar- tion to initiation. In patients with known second- or third-de-
dia and/or AV block. Table 3 lists these medications. Other gree heart block without pacemakers, nodal-blocking agents
mechanisms for consideration include drug overdose, drug should be avoided. Medications should also be adjusted for

PSAP 2022 Book 1 • Cardiology 42 Drug-Induced Cardiovascular Disease


Table 4. Drug-Specific Management of Severe Bradycardia/AV Nodal Blockade

Medication Treatment Options General Approach

β-Blocker/ GI decontamination with activated charcoal Severely symptomatic:


non-DHP CCBs Supportive therapies • Initiate all treatments simultaneously
• IV fluid bolus Mildly symptomatic:
• Calcium chloride IV (up to 3 g total) • Initiate in succession, depending on
• Vasopressors PRN response
HDIE • Atropine + IV fluid bolus
• Regular insulin 1 unit/kg bolus + 0.5- to 1-unit/kg/hr • Glucagon
infusion • Calcium chloride IV
• PRN dextrose 50% 50 mL • Vasopressors
Glucagon 5 mg IV push, 2- to 5-mg/hr infusions • HDIE
Refractory shock: Consider lipid emulsion 20% 100-mL bolus • Lipid emulsions

Digoxin Digoxin-immune Fab (antigen-binding fragments) (DigiFab/ Fab administered to patients with life-
Digibind) threatening or hemodynamically
• Dosing based on acute vs. chronic ingestion unstable arrhythmias, hyperkalemia,
AND evidence of end-organ dysfunction from
• Steady-state serum digoxin concentration hypoperfusion (e.g., renal failure)
OR
• Number of tablets ingested

CCB = non-dihydropyridine calcium channel blocker; HDIE = high-dose insulin/euglycemia; IV = intravenous(ly); PRN = as needed.
Information from: Schult RF, Acquisto NM, Toxicology of cardiovascular drugs. In: Jackevicius C, Patterson JH, eds. Cardiology Self-
Assessment Program, 2020 Book 1. Critical Care Cardiology. American College of Clinical Pharmacy, 2020:67-92; Oliphant CS.
Drug-induced cardiovascular disease and drugs to avoid in CV disease. In: Updates in Therapeutics®: Cardiology Pharmacy
Preparatory Review Course. American College of Clinical Pharmacy 2018:273-300.

renal and hepatic impairment and applicable drug interac- stimulate the sympathetic nervous system (SNS) can cause
tions to ensure accumulation does not occur. these supraventricular arrhythmias (Table 5). Because of
Symptomatic bradyarrhythmias can be mitigated by the uncommon nature of supraventricular arrhythmias, pre-
decreasing or discontinuing the causative agent. If symp- ventive measures are unclear. Management should include
tomatic relief is not provided with this measure or symp-
toms require hospitalization, a temporary pacemaker may
be required while awaiting drug clearance. If there is underly- Table 5. Medications Causing AF or Atrial Flutter and
ing dysfunction of the AV node, permanent pacemaker place- Mechanism
ment may be warranted. In severe cases, atropine may be
administered at a dose of 1 mg every 3–5 minutes (maximum Medication Mechanism

dose 3 mg). If atropine is ineffective, a continuous infusion of Adenosine Shorten atrial


epinephrine intravenously should be initiated at a dose refractory period
of 2–10 mcg/minute or dopamine intravenously at 2–10 Albuterol, alcohol, dobutamine, SNS stimulation
mcg/kg/minute to maintain adequate blood pressures. Table dopamine, theophylline
4 summarizes drug overdose–specific treatments.
Levothyroxine, liothyronine, Hyperthyroidism
thyroid, amiodarone
Atrial Fibrillation/Atrial Flutter Ivabradine Unknown
Atrial fibrillation is a rapid irregularly irregular supraventricu- Bisphosphonates
lar arrhythmia caused by multiple ectopic impulses originat-
ing from the atria and not the SA node. Electrocardiographic SNS= sympathetic nervous system
characteristics include an absence of P waves. In contrast, Information from: Oliphant CS. Drug-induced cardiovascular
the ECG pattern of AF has discernable P waves that produce disease and drugs to avoid in CV disease. In: Updates in
a sawtooth-like pattern. Therapeutics: Cardiology Pharmacy Preparatory Review
Course. American College of Clinical Pharmacy,
Atrial fibrillation and flutter are not typically drug induced, 2018:273-300.
but medications that shorten the atrial refractory period or

PSAP 2022 Book 1 • Cardiology 43 Drug-Induced Cardiovascular Disease


discontinuation of potentially causative agents. If the arrhyth- Heart failure with reduced EF is a risk factor for VT; there-
mia persists, patients should be treated according to current fore, class Ic antiarrhythmics are contraindicated. Class Ia
AHA or ACC guideline recommendations. and Ic should be avoided in coronary artery disease. Digoxin
should be dose adjusted in patients with renal impairment
Ventricular Arrhythmias
and closely monitored during acute kidney injury. Concen-
Monomorphic VT trations above 2 ng/mL are considered toxic. Monitor for
Monomorphic VT originates from ectopic focus in the ven- potential drug-drug interactions when initiating new antiar-
tricles causing ventricular depolarization. Monomorphic VT rhythmics. Maintain normal electrolyte concentrations and
is defined by a regular wide QRS complex with a rate greater consider closer monitoring during periods of GI illnesses
than 100 beats/minute, with a uniform and stable morphol- such as excessive vomiting or diarrhea.
ogy. Monomorphic VT is described as sustained or unsus- If VT occurs, discontinue the potential offending medica-
tained, with unsustained monomorphic VT terminating tion. Direct cardioversion may be necessary to restore sinus
within less than 30 seconds. Monomorphic VT can be related rhythm if the patient is hemodynamically unstable. If the
to medication use or other CV or metabolic causes. Non– patient is hemodynamically stable, management includes
medication-related risk factors include structural heart dis- intravenous antiarrhythmics from several classes such as
ease, depressed left ventricular function (EF less than 40%), adenosine, procainamide, amiodarone, or sotalol.
and electrolyte depletion, particularly magnesium, potas-
sium, and calcium. Diagnosis of drug-induced VT requires Torsades de Pointes
careful consideration of the patient’s medication history, Torsades de pointes (TdP) or polymorphic VT comes from the
elimination of CV pathophysiology, and differentiation from French phrase meaning “twisting of the points.” Torsades de
previous episodes, if any. Vaughan Williams class Ic anti- pointes is the result of prolongation of the ventricular action
arrhythmics are commonly implicated in drug-induced VT potential, evidenced on ECG as QTc prolongation. Torsades de
because their inhibition of sodium channels results in reduc- pointes is potentially life threatening, and quick recognition
tion in ventricular conductivity (Table 6). is critical to prevent deterioration to ventricular fibrillation.
Torsades de pointes can be congenital or acquired. Acquired
TdP is often a result of medications that lengthen the rela-
Table 6. Medications Associated with Monomorphic tive refractory period. Lengthening of the refractory period
Ventricular Tachycardia can occur with decreases in outward current or increases in
inward current during repolarization.
Medication Mechanism There are several risk factors for TdP (Box 2). Prolongation
Digoxin Sodium-potassium-adenosine of the QTc is a crucial step in TdP development. Onset can
triphosphate pump inhibition, be variable, coinciding with the peak of the drug, or delayed
resulting in increased as the drug accumulates. The risk of TdP increases when the
intracellular calcium QTc interval is prolonged over 500 milliseconds. Table 7 lists
Amiodarone, disopyramide, Sodium channel inhibition commonly associated medications.
flecainide, procainamide,
propafenone
Amphetamines, cocaine. SNS stimulation
dobutamine, dopamine,
Box 2. Risk Factors for Drug-Induced
terbutaline
Torsades de Pointes
Milrinone, theophylline PDE inhibition preventing
breakdown of cAMP,
• Additive effect of > 1 agent used in combination
causing increased
• Advanced age
• Bradycardia
intracellular calcium and • Depressed left ventricular ejection fraction
ventricular ectopic activity • Elevated drug concentration (alterations in metabolism
Adenosine, regadenoson SNS stimulation or excretion)
Myocardial ischemia • Female sex
• Hypomagnesemia, hypokalemia
PDE = phosphodiesterase; SNS = sympathetic nervous • QTc > 500 ms
system.
ms = milliseconds.
Information from: Oliphant CS. Drug-induced cardiovascular
Information from: Wiggins BS, Lehner AL. Drug-induced car-
disease and drugs to avoid in CV disease. In: Updates in diovascular disease. In: Jackevicius C, Patterson JH, eds.
Therapeutics: Cardiology Pharmacy Preparatory Review Cardiology Self-Assessment Program, 2020 Book 1. Critical
Course. American College of Clinical Pharmacy, 2018: Care Cardiology. American College of Clinical Pharmacy,
273-300. 2020:93-119.

PSAP 2022 Book 1 • Cardiology 44 Drug-Induced Cardiovascular Disease


Table 7. Medications with Known Risk of Drug-Induced Torsades de Pointes

Drug Class Agents

Antiarrhythmics Amiodarone, disopyramide, dofetilide, dronedarone, flecainide, ibutilide, procainamide,


propafenone, quinidine, sotalol

Antidepressants Citalopram, escitalopram

Antiemetics Chlorpromazine, domperidone, droperidol, ondansetron

Antimicrobials Azithromycin, clarithromycin, ciprofloxacin, erythromycin, fluconazole, levofloxacin,


moxifloxacin, pentamidine

Antipsychotics Haloperidol, thioridazine

Chemotherapeutics Arsenic trioxide, oxaliplatin, vandetanib

Opioid agonists Methadone

Information from: Wiggins BS, Lehner AL. Drug-induced cardiovascular disease. In: Jackevicius C, Patterson JH, eds. Cardiology
Self-Assessment Program, 2020 Book 1. Critical Care Cardiology. American College of Clinical Pharmacy, 2020:93-119.

Torsades de pointes can be prevented through early recog- Volume Retention


nition of risk factors and careful monitoring. A baseline ECG Arterial blood pressure is regulated under normal conditions
should be obtained, when feasible, and a repeat ECG should by the RAAS in the kidneys. Through signaling pathways, hor-
be obtained when the medication has reached steady state. mone release, and sensed changed in pressure and flow, the
Use of prolonging agents should be avoided if the baseline body can adapt by regulating intravascular volume. Inter-
ECG reveals a QTc interval greater than 450 milliseconds. action with the RAAS leading to volume retention occurs
Contributing medications should be discontinued if the QTc for several common classes. These adverse effects can be
exceeds 500 milliseconds. A full medication profile review unwanted and lead to harmful effects in certain patient pop-
helps elucidate synergistic effects of several QTc-prolongat- ulations. Commonly implicated drug classes include NSAIDs,
ing medications. Normal potassium and magnesium con- sex hormones, and corticosteroids.
centrations should be maintained, and closer monitoring
during GI illnesses, such as vomiting or diarrhea, should be Nonsteroidal Anti-inflammatory Drugs
considered. The availability of several NSAIDs OTC makes this drug class
Management of TdP relies on distinguishing polymorphic a common offender of drug-induced hypertension. Nonsteroi-
VT from monomorphic morphology. Unstable patients should dal anti-inflammatory drugs inhibit cyclooxygenase (COX)-1
be treated with defibrillation. Administration of intravenous and COX-2, causing a reduction in prostaglandin synthesis.
magnesium has been reported to terminate TdP and is admin- Prostaglandins have vasodilatory and natriuretic effects.
istered at a dose of 1–2 g over 15 minutes. Figure 1 summa- Nonsteroidal anti-inflammatory drugs increase sodium and
rizes TdP management. water retention and activate the RAAS. Prolonged NSAID use
can result in an increase in blood pressure by an average of
Hypertension 5 mm Hg. Naproxen, piroxicam, and ibuprofen have the high-
Hypertension was the leading cause of death worldwide in est increases (Lovell 2017). However, low-dose aspirin has
2010. Almost one-half of all U.S. adults have hypertension, almost no effect on blood pressure.
and only one-fourth of cases are under control (CDC 2020). Nonsteroidal anti-inflammatory drugs also reduce the effi-
Drug-induced hypertension can be caused by use or with- cacy of RAAS inhibitor agents, β-blockers, and diuretics used
drawal of a medication and is often overlooked as a poten- for the treatment of preexisting hypertension. Alternative
tial cause of resistant hypertension. Hypertension is often agents such as calcium channel blockers and centrally act-
preventable and reversible. A careful medication history ing agents can be used in patients who cannot discontinue
including OTC and herbal supplements is necessary to iden- NSAID therapy.
tify potential sources of drug-induced hypertension. Causes
include volume retention, sympathomimetic activation, and Sex Hormones
direct vasoconstriction. In addition, abrupt withdrawal of
Oral contraceptives containing estrogen can activate the
β-blockers and centrally acting α-agonists can cause rebound
RAAS by increasing the production of angiotensinogen,
hypertension.
resulting in volume retention. Higher doses of estrogen than

PSAP 2022 Book 1 • Cardiology 45 Drug-Induced Cardiovascular Disease


Torsades de pointes

Discontinue offending agent(s)


Correct electrolytes (K+, Mg2+, Ca2+)

Hemodynamically Hemodynamically
unstable stable

Defibrillation Magnesium sulfate


1–2 g IV over 15 min

TdP recurrence

TdP with bradycardia TdP with no


or precipitated by bradycardia or does
pauses in rhythm not appear to be
precipitated by
pauses in the rhythm

Isoproterenol 2- to 10-mcg/min
continuous IV infusion
OR TdP recurrence Defibrillation
Temporary transvenous
overdrive pacing

Figure 1. Management of drug-induced torsades de pointes.


IV = intravenous(ly); TdP = torsades de pointes.
Information from: Tisdale JE. Ventricular arrhythmias. In: Tisdale JE, Miller DA, eds. Drug-Induced Diseases: Prevention, Detection,
and Management. American Society of Health-System Pharmacists, 2018:523-67.

previously used have a more pronounced effect; however, sodium and water retention. Testosterone is a known modu-
increases in blood pressure can occur with newer, lower dos- lator and regulator of metabolic functions, including choles-
ing strategies. Hormone therapy from contraception supplies terol synthesis and heart function. Testosterone deficiency
higher concentrations of estrogen and progestin than nor- leads to metabolic syndrome, resulting in hypertension. How-
mally detected. In contrast, doses of exogenous hormones ever, replacement in hypogonadal men has been shown to
given to postmenopausal women for the prevention of meno- decrease blood pressure by an average of 23 mm Hg systolic
pausal symptoms are lower than what is found in vivo during and 16 mm Hg diastolic (Francomano 2014).
menopause. Although estrogen replacement therapy in both
populations of women results in a higher risk of CV events, Corticosteroids
drug-induced hypertension is uncommon in postmenopausal Corticosteroids include both mineralocorticoids and glu-
women. The incidence of hypertension in patients taking oral cocorticoids, with mineralocorticoid activity implicated in
contraceptives is about 5% (Lovell 2017). drug-induced hypertension through sodium and water reten-
Testosterone causes fluid retention through androgen tion. Steroids that have greater mineralocorticoid activ-
receptor agonism, which stimulates erythropoietin. Increases ity (i.e., hydrocortisone and fludrocortisone) have a greater
in blood volume through polycythemia lead to edema from effect on blood pressure. However, other steroids can have

PSAP 2022 Book 1 • Cardiology 46 Drug-Induced Cardiovascular Disease


mineralocorticoid activity at higher doses (7.5 mg/day or receptors in the heart. The resultant effect is hypertension
more of prednisone equivalents). Twenty percent of patients from peripheral vasoconstriction, tachycardia, and increased
who use corticosteroids have hypertension, with elevations contractility. Cocaine causes a dose-dependent increase in
of as much as 15 mm Hg at cortisol doses of 80–200 mg blood pressure as well as other effects depending on route of
(Kassel 2015). Some studies suggest that CV risk is more pro- administration (Lovell 2017).
nounced at higher steroid doses than during long treatment
durations (Lovell 2017). Antidepressants and Antipsychotics
Several antidepressants and antipsychotics have been iden-
Sympathomimetic Activation
tified as potentially contributing to drug-induced hyperten-
Decongestants sion. Bupropion, a dopamine reuptake inhibitor, is associated
Pseudoephedrine and phenylephrine are both available OTC with elevations in supine blood pressure while causing
for nasal congestion. Pseudoephedrine and phenylephrine orthostatic hypotension. Risk of hypertension is increased
work by activating the SNS through α1-adrenergic receptors, in patients using transdermal nicotine patches for smoking
causing peripheral vasoconstriction. Phenylephrine doses of cessation.
45 mg can increase systolic pressures by 20 mm Hg (Lovell Tricyclic antidepressants were some of the first antidepres-
2017). Many formulations co-formulated with acetamino- sants to be used. Each drug in this class works similarly by pre-
phen pose an additional concern because coadministration venting reuptake of serotonin and norepinephrine. However,
increases the bioavailability of phenylephrine. Pseudoephed- tricyclic antidepressants have unintended adverse effects
rine also causes a dose-dependent increase in blood pres- because of receptor selectivity. Some members of this class
sure, though the clinical implications are unclear. Increases have anticholinergic, antihistaminic, and α1-adrengeric recep-
in both normotensive patients and patients with hypertension tor activity. Tricyclic antidepressants should be reserved for
were less than 2 mm Hg. Decongestants should be used with patients who cannot be treated with newer-generation antide-
caution in patients with hypertension; however, they are gen- pressants (Kassel 2015).
erally considered safe. Monoamine oxidase inhibitors are associated with eleva-
tions in blood pressure with ingestion of tyramine-rich foods.
Psychostimulants Possible mechanisms by which hypertension occurs include
Sympathetic stimulation from medications used for atten- metabolism to amphetamine. Monoamine oxidase inhibi-
tion-deficit/hyperactivity disorder increases the norepi- tors should only be used in patients who are refractory to or
nephrine in the presynaptic terminal, activating adrenergic unable to take first-line therapies (Kassel 2015).
receptors, leading to vasoconstriction. Data in the pediat- Venlafaxine, a serotonin and norepinephrine reuptake
ric population show that stimulants increase diastolic blood inhibitor, causes hypertension at doses greater than 300 mg/
pressure and heart rate, but not systolic blood pressure (Kas- day. Doses should be limited in patients and additional agents
sel 2015). The AHA recommends blood pressure and heart added on if symptoms are uncontrolled at maximum doses.
rate monitoring within 1–3 months after initiation. Follow-up Venlafaxine’s enantiomer, desvenlafaxine, also causes eleva-
monitoring should occur every 6–12 months and more often tions in blood pressure, particularly in those with preexisting
during periods of titration or weaning (Vetter 2008). CVD (Kassel 2015).

Caffeine
Direct Vasoconstriction
Caffeine causes an increase in blood pressure by activat-
Calcineurin inhibitors (CNIs) tacrolimus and cyclosporine
ing the SNS, increasing catecholamine release, and blocking
are immunosuppressive agents used for the prevention of
circulating adenosine, responsible for coronary vasodila-
solid organ transplant rejection together with several other
tion (Lovell 2017). One meta-analysis found that acute con-
conditions. Because there are no alternatives to therapy,
sumption of 200–300 mg of caffeine caused an increase of
adverse effects must be managed. Drug-induced hyperten-
8.14 mm Hg in systolic blood pressure within the first several
sion occurs through several pathways. Calcineurin inhibitors
hours. Drinking caffeine (coffee) for 2 weeks did not appear
decrease the production of nitric oxide, resulting in inhibi-
to raise participants’ blood pressure (Mesas 2011). Long-term
tion of vasodilation. In addition, SNS stimulation causes
effects of drinking caffeine are unknown, but drinking caf-
sodium retention. It appears that cyclosporine more com-
feine was not associated with worse outcomes.
monly causes drug-induced hypertension and that effects
are dose related (Lovell 2017). Management of CNI-induced
Cocaine
hypertension can be through dose reduction or addition of
Like other psychostimulants, cocaine increases norepineph-
antihypertensives. Expert opinion suggests reducing the CNI
rine at the synapse, resulting in sympathetic stimulation of
dose by 25% for stage 1 hypertension and 50% for stage 2.
α-adrenergic receptors in the periphery but also β-adrenergic

PSAP 2022 Book 1 • Cardiology 47 Drug-Induced Cardiovascular Disease


Dihydropyridine calcium channel blockers are the preferred such as trazodone. Lowering the dose or discontinuing the
antihypertensive because of their lack of drug interactions medication should be considered in symptomatic patients.
and mechanism. Dihydropyridine calcium channel blockers Tricyclic antidepressant overdose can cause profound hypo-
are particularly beneficial in kidney transplant recipients by tension requiring medical intervention, including intravenous
causing vasodilation in both the afferent and efferent arteri- fluid boluses and use of vasopressors. Lowering of antihyper-
oles (Claus 2018). tensives may also be required to allow the patient to tolerate
the hypotension associated with antidepressant therapy.
Miscellaneous/Other
Antiparkinsonian agents work by increasing the amount
Erythropoiesis-stimulating agents epoetin alfa and darbep- of circulating dopamine. Hypotension can occur through
oetin alfa are FDA approved to treat anemia for a variety of vasodilation by dopamine receptors in the mesenteric and
pathologies, including chronic kidney disease and HIV, and as renal circulation, shift of norepinephrine from receptor sites,
an adjunct to chemotherapy. Hypertension occurs in about decreased sympathetic outflow, and inhibition of renin and
20%–30% of patients, developing in the subacute treatment aldosterone secretion. Several agents exist in this class,
phase. Several different mechanisms exist, including increase including bromocriptine, carbidopa/levodopa, entacapone,
in blood viscosity, RAAS activation, and increased vasocon- pergolide, pramipexole, rasagiline, ropinirole, selegiline, and
striction through decreased nitric oxide synthesis. If discon- tolcapone. No specific therapies for hypotension are asso-
tinuing the medication is not feasible, several strategies can ciated with antiparkinsonian medications. Nonpharmaco-
be used to lower the blood pressure. In patients with chronic logic strategies that may be beneficial include increasing
kidney disease, volume removal may prove helpful. Addition fluid and salt intake, tilting the head of bed during nighttime
of diuretics and antihypertensives to control the blood pres- sleep, using compression stockings or abdominal bandages,
sure elevation can also be considered. Patients taking eryth- and performing physical movements such as leg crossing
ropoiesis-stimulating agents should have their doses lowered or squatting (Sánchez-Ferro 2013). If these do not produce
or discontinued when no longer indicated. Blood pressure the desired effect, pharmacologic agents can be used. Flud-
should be monitored throughout therapy (Claus 2018). rocortisone, pyridostigmine, midodrine, and droxidopa are
all used to improve hypotension associated with antipar-
Hypotension kinsonian therapy. Fludrocortisone works by expanding vol-
Hypotension, defined as blood pressure less than 90/60 mm ume through renal sodium reabsorption. Pyridostigmine
Hg, can often result from over-intensification of an antihyper- inhibits cholinesterase, improving transmission of acetyl-
tensive regimen or as an unintended adverse effect because choline signaling. Midodrine is an α1-agonist that increases
of receptor selectivity. Hypotension is potentially problematic systemic vascular resistance. Finally, droxidopa is a syn-
in older adult patients because it can lead to syncope, falls thetic norepinephrine precursor that can used in the setting
and injury, or hypoperfusion of vital organ systems; there- of severe orthostasis from autonomic disorders. Of impor-
fore, it should be prevented. Hypotension can be categorized tance, although these medications can improve orthostatic
as acute, chronic, or orthostatic. Chronic implies the blood hypotension, they can worsen supine hypertension. Fre-
pressure is consistently low despite the patient’s position. quent monitoring is recommended to avoid overcorrection
Orthostatic describes a reduction in blood pressure when the of hypotension.
patient stands up. Hypotension can also be the result of auto- Endothelin (ET) receptor antagonists are used to treat pul-
nomic failure. monary arterial hypertension. Inhibition of ET-1 causes vaso-
Hypotension risk is additive when several antihyperten- dilation in smooth muscle at receptor targets ETA and ETB.
sives are prescribed. Other risk factors include alcohol use, Available agents include nonselective bosentan and maci-
dehydration and hot weather, hot tubs, bath or shower use, tentan and the selective ETA inhibitor ambrisentan. Because
and prolonged sitting or lying down. Several other agents of vasodilation, hypotension can result. The risk appears to
can induce hypotension unrelated to antihypertension be low but can be synergistic if other antihypertensives are
mechanisms. added.
Intravenous amiodarone, a common antiarrhythmic, is sus-
pended in polysorbate-80 and benzyl alcohol. Polysorbate-80
Myocardial Ischemia
has potent vasodilatory and negative inotropic effects. Ben-
zyl alcohol also causes hypotension. It is recommended to Several mechanisms for medication-induced myocardial
administer an amiodarone 150-mg bolus over 10 minutes to ischemia exist. Myocardial ischemia results from an oxy-
prevent hypotension. The infusion can be slowed further if gen supply and demand mismatch. Commonly, ischemia
hypotension occurs. can occur from the development of atherosclerotic disease,
Tricyclic antidepressants have activity at unintended though coronary vasospasm or drug withdrawal can also
receptors, causing hypotension. Hypotension is common with lead to infarction. Ischemia can occur acutely, which may
imipramine but is also reported with other antidepressants be directly attributable to a drug. Acute coronary syndromes

PSAP 2022 Book 1 • Cardiology 48 Drug-Induced Cardiovascular Disease


that are associated with longer-term therapy are more chal- protective against MI (Llibre 2016). On the basis of current
lenging to prove a direct correlation with; however, some ther- evidence, it is reasonable to avoid use of abacavir in patients
apies have been noted to increase the risk of atherosclerotic at risk of CVD. Although some regimens are unavoidable
coronary artery disease and its associated morbidity. because of resistance patterns, other combination therapies
should be considered in treatment-naive at-risk patients.
HIV Medications
Antiretroviral therapy for HIV infection has prolonged the Nonsteroidal Anti-inflammatory Drugs
lives of many with HIV infection. Several medications have
Nonsteroidal anti-inflammatory drugs are widely available
been linked to an increased risk of CVD, mainly limited to old-
and commonly prescribed for pain management. Neverthe-
er-generation medications from the protease inhibitor and
less, they have been associated with an increased risk of
nucleoside reverse transcriptase inhibitor classes. An analy-
CV events. This risk has garnered attention in recent years
sis of Veterans Affairs registry data showed that patients with
because of the removal of two selective COX-2 inhibitors from
HIV-positive infection had a 48% increased risk of MI (Freiberg
the U.S. market, rofecoxib and valdecoxib. Overall, major vas-
2013). Both HIV and antiretroviral therapy can contribute to
cular events are more frequently seen with the use of NSAIDs
CVD. Human immunodeficiency virus infection itself alters
compared to placebo, regardless of COX selectivity. Cardio-
immune function, causes chronic inflammation, and predis-
vascular risk seems to be lower for nonselective NSAIDs
poses patients to dyslipidemia and hypercoagulable states.
like naproxen and ibuprofen but is still higher than seen in
Antiretroviral therapy has several long-term adverse effects,
non-users. Additionally, available COX-2 selective agents
including insulin resistance, dyslipidemia, and lipodystrophy
such as celecoxib demonstrate increased rates of vascular
(Overton 2014). Protease inhibitors cause increases in serum
events compared to nonselective agents. The AHA issued a
cholesterol and triglycerides. Increases in cholesterol of
recommendation in 2014 stating that analgesic agents with
30%–40% and triglycerides of 200%–300% can occur within
lower risk (e.g., acetaminophen, tramadol) should be used
the first week of therapy. In addition, after 2 years of therapy,
first line in the treatment of musculoskeletal pain symptoms
more than 50% of patients taking protease inhibitors will have
in patients with known CVD or risk factors for CVD. Naproxen
hypercholesterolemia or hypertriglyceridemia (Penzak 2002).
is preferred if the other agents are ineffective, according to
Abacavir, a nucleoside reverse transcriptase inhibitor, can
a meta-analysis of randomized controlled trials showing no
cause CVD. Recent exposure to abacavir was associated
increase in risk (Patrono 2014).
with increased rates of acute MI. Several other cohort stud-
ies failed to replicate this finding. Two more recent studies Heart Failure
have added more data to the debate. The first, NA-ACCORD,
Heart failure is estimated to affect 6.2 million Americans.
showed that treatment-naive patients initiated on abacavir-
Heart failure is the primary diagnosis for hospital discharge
containing regimens had an increased risk of acute MI. The
for 1 million and secondary diagnosis for 2 million hospital-
second study of a Swiss cohort found that recent exposure
izations per year (Hollenberg 2019). Patients with HF are pre-
to abacavir in the past 6 months was associated with an
scribed an average of seven prescription drugs with complex
increased risk of acute MI; however, current abacavir use was
dosing regimens (Page 2016). Polypharmacy, usually defined

Table 8. Medications That May Cause or Exacerbate HF

Medication Cause HF Exacerbate HF Mechanism Onset

α1-Antagonists: X β1-Receptor stimulation Intermediate to delayed


Doxazosin

Amphotericin B X Unknown Unknown

Anesthetics: Immediate

Dexmedetomidine X α 2-Agonist

Etomidate X Suppression of adrenal


function

Ketamine X Negative inotrope

(continued)

PSAP 2022 Book 1 • Cardiology 49 Drug-Induced Cardiovascular Disease


Table 8. Medications That May Cause or Exacerbate HF  (continued)

Propofol X Negative inotrope,


vasodilation

Anthracyclines: X X Reduced contractility, many Immediate to delayed


Doxorubicin potential mechanisms
Epirubicin
Idarubicin

Antiarrhythmics: X Negative inotrope Immediate to intermediate


Disopyramide
Dronedarone
Flecainide
Lidocaine
Mexiletine
Propafenone

Antidiabetics: Immediate to delayed

Dipeptidyl peptidase-4 X Unknown


inhibitors:
Saxagliptin
Sitagliptin

Thiazolidinediones X Possible calcium channel


blockage

Calcium channel blockers: X Negative inotrope Immediate


Diltiazem
Nifedipine
Verapamil

Cilostazol X PDE-3 inhibitor, resulting in Unknown


arrhythmias

Clozapine X IgE-mediated Intermediate to delayed


hypersensitivity reaction,
calcium channel blockage

COX-2 inhibitors: X Sodium and water retention Immediate


Celecoxib

HER2 receptor inhibitor: X X Antibody-dependent Immediate to delayed


Trastuzumab cytotoxicity

Parkinson disease agents: X Excess serotonin activity Intermediate to delayed


Pramipexole leading to valvular damage
Ropinirole

Tricyclic antidepressants X Negative inotrope, Intermediate to delayed


proarrhythmic

VEGF inhibitors: X Unknown Unknown


Bevacizumab
Sunitinib
Sorafenib
Ponatinib

HER2 = human epidermal growth factor; IgE = immunoglobulin E.


Information from: Page RL II, O’Bryant CL, Cheng D, et al. Drugs that may cause or exacerbate heart failure: a scientific statement
from the American Heart Association [published correction appears in Circulation 2016;134:e261]. Circulation 2016;134:e32-e69;
Hamarneh YN, Tsuyuki RT. Heart failure. In: Tisdale JE, Miller DA, eds. Drug-Induced Diseases: Prevention, Detection and
Management. American Society of Health-System Pharmacists 2018:501-21.

PSAP 2022 Book 1 • Cardiology 50 Drug-Induced Cardiovascular Disease


Armenian SH, Lacchetti C, Barac A et al. Prevention and
Practice Points monitoring of cardiac dysfunction in survivors of adult
• Patients with cancer, especially those with breast cancer, cancers: American Society of Clinical Oncology Clinical
face an elevated risk of cardiomyopathy in their lifetime Practice Guideline. J Clin Oncol 2016 35:893-911.
secondary to first-line therapy with anthracyclines and/or
HER2 inhibitors. Screening for and management of CV risk Barac A, Murtagth G, Carver JR et al. Cardiovascular health
factors can help mitigate overall risk. of patients with cancer and cancer survivors:
• Both bradyarrhythmias and tachyarrhythmias can be in- a roadmap to the next level. J Am Coll Cardio 2015 Jun 30;
duced by the overprescribing of multiple cardiac medica- 65:2739–46.
tions. It is important to recognize which medications will
Blanter JB, Frishman WH. The preventative role of angioten-
exacerbate existing arrhythmias and which can unmask
sin converting enzyme inhibitors/ angiotensin-II receptor
electrophysiologic issues within the heart.
blockers and beta-adrenergic blockers in anthracycline-
• Ventricular arrhythmias such as TdP can occur in the
and trastuzumab-inducted cardiotoxicity. Cardiology in
setting of a congenital prolonged QTc disease or as an
Review 2019;12:256-9.
additive effect from multiple medications. Pharmacists
can help patient care teams with alternatives that are less Brahmer JR, Lacchetti C, Schneider BJ, et al. Management of
prolonging or provide information on the risk associated Immune-Related Adverse Events in Patients Treated With
with certain classes of medications. Immune Checkpoint Inhibitor Therapy: American Society
• The cause of medication-induced hypertension is related to of Clinical Oncology Clinical Practice Guideline. J Clin
several mechanisms, including volume retention, sympa- Oncol. 2018;36(17):1714-1768.
thetic activation, and direct vasoconstriction. Pharmacists
can help identify the offending medications and provide Cardinale D, Colombo A, Sandri M et al. Prevention of high-
reasonable alternatives if they exist. dose chemotherapy-induced cardiotoxicity in high-risk
• Antiretroviral therapy and NSAIDs can increase a patient’s patients by angiotensin-converting enzyme inhibition.
risk of CVD. Combined with risk modification, knowing Circulation 2006;114:2474-81.
the risk of the individual agent can help prevent untoward
ischemic heart disease. Cardinale D, Iacopo F, Cipolla CM. Cardiotoxicity of anthracy-
clines. Front Cardiovasc Med 2020; 7: 26.

Centers for Disease Control. Facts about Hypertension. 2020


as five or more medications daily, places patients at risk of Sept.
more adverse drug reactions and drug-drug interactions
Chen MH, Kerkelä R, Force T. Mechanisms of cardiac dys-
(Masnoon 2017). Medications known to cause or exacerbate
function associated with tyrosine kinase inhibitor cancer
HF are too numerous for individual discussion in this chap- therapeutics. Circulation. 2008 Jul 1;118:84-95.
ter. Table 8 summarizes the agents, and the beginning of the
Claus LW, Saseen JJ. Hypertension. In: In: Tisdale JE, Miller
chapter provides suggestions for additional reading.
DA, eds. Drug-induced diseases: prevention, detection
and management. Bethesda MD: American Society of
CONCLUSION Health-System Pharmacists 2018: 617-30.
Drug-induced CVD can present a challenge for both the gen-
Copeland-Halperin RS, Liu JE, Yu AF. Cardiotoxicity of
eral patient population and patients with cancer. Anticipating HER2-targeted therapies. Curr Opin Cardiol 2019 Jul; 34:
potential adverse events can provide the opportunity to opti- 451-8.
mize therapy through screening. Understanding the mecha-
D’Souza M, Carlson N, Fosbøl E, et al. CHA2DS2-VASc score
nism that causes cardiac diseases and the treatment for it
and risk of thromboembolism and bleeding in patients
is vital to providing comprehensive care. Pharmacists are with atrial fibrillation and recent cancer. Eur J Prev Cardiol.
uniquely positioned to provide drug information to the patient 2018;25(6):651-658.
care team. Identification of drug-induced diseases, therapeu-
Domercant J, Polin N, Jahangir E. Cardio-oncology: a focused
tic treatment, prevention strategies, and patient counseling
review of anthracycline-, human epidermal growth factor
are all essential services in the care of our patients. receptor 2 inhibitor-, and radiation-induced cardiotoxicity
and management. Ochsner J. 2016 Fall;15:250-6.
REFERENCES
Florescu M, Cinteza M, Vinereanu D. Chemotherapy-induced
Aghel N, Delgado DH, Lipton JH. Cardiovascular toxicities cardiotoxicity. Maedica (Bucur) 2013 Mar; 8: 59-67.
of BCR-ABL tyrosine kinase inhibitors in chronic myeloid
leukemia: preventive strategies and cardiovascular Francomano D, Lenzi A, Aversa A. Effects of five-year treat-
surveillance. Vasc Health Risk Manag. 2017;13:293-303. ment with testosterone undecanoate on metabolic and
hormonal parameters in ageing men with metabolic
Alexandre J, Cautela J, Ederhy S, et al. Cardiovascular Toxicity syndrome. Int J Endocrinol. 2014;2014:527470.
Related to Cancer Treatment: A Pragmatic Approach to the
American and European Cardio-Oncology Guidelines. Ganatra S, Sharma A, Shah S, et al. Ibrutinib-Associated Atrial
J Am Heart Assoc. 2020;9:e018403. Fibrillation. JACC Clin Electrophysiol. 2018;4(12):1491-1500

PSAP 2022 Book 1 • Cardiology 51 Drug-Induced Cardiovascular Disease


Georgakopoulos P, Kyriakidis M, Perpinia A, et al. The role National Comprehensive Cancer Network: NCCN Clinical
of metoprolol and enalapril in the prevention of doxorubi- Practice Guideline in Oncology. Survivorship Version 1.
cin-induced cardiotoxicity in lymphoma patients. Anti Can Fort Washington, PA, Nation Comprehensive Cancer
Res 2019;39:5703-7. Network. 2021.

Hollenberg SM, Warner Stevenson L, Ahmad T, et al. 2019 Octavia Y, Tocchetti CG, Gabrielson KL et al. Doxorubi-
ACC Expert Consensus Decision Pathway on Risk Assess- cin-induced cardiomyopathy: from molecular mecha-
ment, Management, and Clinical Trajectory of Patients nisms to therapeutic strategies. J Mol Cell Cardiol 2012
Hospitalized With Heart Failure: A Report of the American June;52:1213-25.
College of Cardiology Solution Set Oversight Committee
[published correction appears in J Am Coll Cardiol. 2020 Overton ET. Metabolic complications of HIV infection and its
Jan 7;75(1):132]. J Am Coll Cardiol. 2019;74:1966-2011. therapies. Top Antivir Med. 2014;22:651-4.

Hu YF, Liu CJ, Chang PM, et al. Incident thromboembolism Page RL 2nd, O’Bryant CL, Cheng D, et al. Drugs That May
and heart failure associated with new-onset atrial fibrilla- Cause or Exacerbate Heart Failure: A Scientific Statement
tion in cancer patients. Int J Cardiol. 2013;165(2):355-357. From the American Heart Association [published correc-
tion appears in Circulation. 2016 Sep 20;134(12):e261].
Imbruvica (ibrutinib). Package insert. Janssen Biotech Inc. Circulation. 2016;134:e32-e69.
July 2019.
Patrono C, Baigent C. Nonsteroidal anti-inflammatory drugs
Jerusalem G, Lancellotti P, Kim SB. HER2+ breast cancer and the heart. Circulation. 2014 Feb 25;129(8):907-16.
treatment and cardiotoxicity: monitoring and manage-
ment. Breast Cancer Res Treat. 2019;177(2):237-250. Penzak SR, Chuck SK. Management of protease inhibitor-­
associated hyperlipidemia. Am J Cardiovasc Drugs. 2002;
Koutsoukis A, Ntalianis A, Repasos E, et al. Cardio-oncology: 2:91-106.
A Focus on Cardiotoxicity. Eur Cardiol. 2018;13:64-69.
Sánchez-Ferro A, Benito-León J, Gómez-Esteban JC. The
Lee M, Chung WB, Lee JE, et al. Candesartan and carvedilol for management of orthostatic hypotension in Parkinson’s
primary prevention of subclinical cardiotoxicity in breast disease. Front Neurol. 2013;4:64. Published 2013 Jun 10.
cancer patients without a cardiovascular risk treated with
doxorubicin. Cancer Med. 2021;10(12):3964-3973. Słowik A, Jagielski P, Potocki P, et al. Anthracycline-induced
cardiotoxicity prevention with angiotensin-converting
Leong DP, Caron F, Hillis C, et al. The risk of atrial fibrillation enzyme inhibitor ramipril in women with low-risk breast
with ibrutinib use: a systematic review and meta-analysis. cancer: results of a prospective randomized study. Kardiol
Blood 2016 Jul 7;128:138-40. Pol. 2020;78(2):131-137.

Lin H, Liang G, Wu Y, Chen L. Protective Effects of ACEI/ARB Swain SM, Ewer MS, Cortés J, et al. Cardiac tolerability of
on Left Ventricular Function in Anthracycline-Induced pertuzumab plus trastuzumab plus docetaxel in patients
Chronic Cardiotoxicity: A Meta-Analysis of Randomized with HER2-positive metastatic breast cancer in CLEOP-
Controlled Trials. Cardiology. 2021;146(4):469-480. ATRA: a randomized, double-blind, placebo-controlled
phase III study. Oncologist. 2013;18:257-64.
Llibre JM, Hill A. Abacavir and cardiovascular disease: A
critical look at the data. Antiviral Res. 2016 Aug;132:116-21. Vetter VL, Elia J, Erickson C, et al.; American Heart Asso-
ciation Council on Cardiovascular Disease in the Young
Lovell AR, Ernst ME. Drug-Induced Hypertension: Focus Congenital Cardiac Defects Committee; American Heart
on Mechanisms and Management. Curr Hypertens Rep. Association Council on Cardiovascular Nursing. Cardio-
2017;19(5):39. vascular monitoring of children and adolescents with
Masnoon N, Shakib S, Kalisch-Ellett L, Caughey GE. What is heart disease receiving medications for attention deficit/
polypharmacy? A systematic review of definitions. BMC hyperactivity disorder [corrected]: a scientific statement
Geriatr. 2017;17(1):230. Published 2017 Oct 10. from the American Heart Association Council on Cardio-
vascular Disease in the Young Congenital Cardiac Defects
Mesas AE, Leon-Muñoz LM, Rodriguez-Artalejo F, et al. The Committee and the Council on Cardiovascular Nursing.
effect of coffee on blood pressure and cardiovascular dis- Circulation. 2008 May 6;117:2407-23.
ease in hypertensive individuals: a systematic review and
meta-analysis. Am J Clin Nutr. 2011 Oct;94:1113-26. Yeh ETH, Bickford CL. Cardiovascular complications of
cancer therapy. J Am Coll Cardiol 2009:53:2231-47.
Michel L, Rassaf T, Totzeck M. Cardiotoxicity from
immune checkpoint inhibitors. Int J Cardiol Heart Vasc.
2019;25:100420. Published 2019 Sep 7.

More L, Patell, R, Asnani, A. Ibrutinib-Associated Cardiotoxic-


ity. American College of Cardiology. 2020 Jan 21.

PSAP 2022 Book 1 • Cardiology 52 Drug-Induced Cardiovascular Disease


Self-Assessment Questions
Questions 16 and 17 pertain to the following case. C. Her blood pressure is not at goal; amlodipine should
J.E. is a 55-year-old woman diagnosed with breast cancer. be increased to 10 mg daily and her blood pressure
She will soon undergo chemotherapy with doxorubicin. Her rechecked in 2 weeks.
anticipated cumulative dose will be 300 mg/m2. Her medical D. Her blood pressure is not at goal; amlodipine should
history includes hypertension, for which she takes hydrochlo- be changed to lisinopril because this will prevent
rothiazide 25 mg daily. progression to HF.
16. Which one of the following is best to recommend to 20. A 29-year-old man (weight 90 kg) with metastatic mela-
decrease J.E.’s risk of cardiotoxicity? noma presents to the ED with chest pain, shortness of
A. Use standard formulation of doxorubicin. breath, and lower extremity swelling. His home drugs
B. Change antihypertensive to lisinopril 10 mg daily. include ipilimumab 270 mg intravenously every 3 weeks.
C. Change antihypertensive to metoprolol tartrate His last dose was over 2 weeks ago. The consulting car-
25 mg twice daily. diology team diagnoses clinically suspected myocardi-
D. Administer dexrazoxane before doxorubicin infusion. tis and admits him for observation and management of
edema. Which one of the following is best to recommend
17. Four months after completing the doxorubicin infusions,
regarding this patient’s anticancer treatment?
J.E. has shortness of breath and is evaluated by a car-
diologist for new-onset heart failure (HF). Her new left A. Permanently discontinue ipilimumab therapy.
ventricular ejection fraction (LVEF) is 35%. Given her new B. Reinitiate ipilimumab therapy as inpatient because
diagnosis of HF with reduced EF, which one of the follow- his next dose is due soon.
ing is best to recommend for J.E.? C. Hold next ipilimumab dose and reinitiate after
hospital discharge, after symptoms resolve.
A. Discontinue hydrochlorothiazide and initiate
D. Initiate corticosteroid taper and resume ipilimumab
guideline-directed medical therapy.
after completion.
B. Continue hydrochlorothiazide.
C. Change hydrochlorothiazide to furosemide. 21. A 78-year-old man presents to the ED with syncope. On
D. Refer her to a cardio-oncologist for further presentation, his heart rate is 45 beats/minute. His home
evaluation and initiate guideline-directed medical drugs include amiodarone 200 mg daily, rivaroxaban
therapy. 20 mg daily, metoprolol succinate 50 mg daily, and levothy-
roxine 88 mcg daily. The attending physician asks which
18. A 76-year-old man will be initiated on ponatinib for chronic
medication most likely caused his symptomatic bradycar-
myeloid leukemia. Which one of the following is best to
dia. Which one of the following is the best response?
recommend for this patient before initiating therapy?
A. Amiodarone and metoprolol can cause bradycardia.
A. Cardio-oncologic evaluation and ankle-brachial
B. Metoprolol is likely the sole cause of bradycardia.
index with lower extremity Doppler
C. Levothyroxine has been associated with
B. Cardio-oncologic evaluation with echocardiography
bradycardia.
C. Serum troponin
D. Rivaroxaban could be contributing to a hemorrhage-
D. Nuclear stress test
associated bradycardia.
19. A 54-year-old woman was recently diagnosed with met-
22. A 65-year-old man underwent an elective total knee
astatic pancreatic neuroendocrine tumors, and the team
replacement. His medical history includes hyperten-
is discussing treatments with her. She has a history of
sion, depression, and benign prostatic hyperplasia. His
hypertension and takes amlodipine 5 mg daily at home.
home drugs include lisinopril 10 mg daily, doxazosin
Her vital signs today include heart rate 69 beats/minute
4 mg daily, sertraline 100 mg daily, and metoprolol 50 mg
and blood pressure 127/79 mm Hg. The patient and the
twice daily. On the morning of surgery, he forgot to take
team want to initiate sunitinib. Which one of the follow-
his home drugs. His blood pressure after the operation is
ing is best to recommend for this patient?
195/94 mm Hg. Which one of the following is most likely
A. Her blood pressure is at goal; no change is contributing to this patient’s hypertension?
necessary. She can return to the clinic for a blood
A. Lisinopril
pressure check 4 weeks after initiation of therapy.
B. Doxazosin
B. Her blood pressure is at goal; she should start
C. Sertraline
home blood pressure monitoring for the duration of
D. Metoprolol
sunitinib therapy.

PSAP 2022 Book 1 • Cardiology 53 Drug-Induced Cardiovascular Disease


23. During rounds with your patient care team, the attending He is diagnosed with COVID-19 pneumonia, and the
physician is discussing drugs that have been removed physician would also like to treat him for community-
from the market because of safety concerns. The sub- acquired pneumonia. His medical history includes
ject of rofecoxib is mentioned. Which one of the follow- severe mitral regurgitation, depression, and hyperten-
ing best evaluates the current evidence with respect to sion. At home, he takes escitalopram 20 mg daily, furo-
cardiovascular disease (CVD) and NSAID use? semide 80 mg daily, and valsartan 160 mg daily. The
physician writes a prescription for azithromycin 500 mg
A. Rofecoxib was removed from the market because of
daily today, then 250 mg daily and cefpodoxime 200 mg
increased risk of bleeding.
twice daily. As you review the medication orders for the
B. Risk of ischemic heart disease is lowest with
outpatient clinic, which one of the following is best to
selective COX-2 inhibitors like celecoxib.
recommend for this patient?
C. Selective COX-2 inhibitors are the only NSAIDs that
cause hypertension. A. Continue azithromycin; discontinue escitalopram
D. Naproxen is associated with increased blood while on antibiotics.
pressure. B. Continue azithromycin and cefpodoxime; monitor
QTc daily while on antibiotics.
24. An 87-year-old woman (weight 50 kg) is admitted to
C. Discontinue azithromycin and cefpodoxime; initiate
the hospital after experiencing altered mental status at
levofloxacin 750 mg daily.
home. On arrival at the ED, she is dehydrated with dry
D. Discontinue azithromycin; initiate doxycycline
mucous membranes and an SCr of 4.5 mg/dL (base-
100 mg twice daily.
line 1.2 mg/dL) and a potassium of 4.1 mEq/L. She has
a medical history of atrial fibrillation on digoxin 125 mcg 27. You are rounding with an internal medicine team and
daily and rivaroxaban 15 mg with dinner. The patient’s reviewing home drugs for a man who was admitted with
blood pressure is 100/76 mm Hg, heart rate is 76 beats/ new-onset HF. The patient’s home drugs include nifed-
minute, and ECG is normal. A routine digoxin concentra- ipine extended release 90 mg daily, metformin 500 mg
tion is 2.2 ng/mL. The patient has no concerns for visual twice daily, lisinopril 20 mg daily, and clozapine 100 mg
disturbances. Which one of the following is best to rec- daily. The team wants to know which of his home drugs
ommend for this patient? may be contributing to his HF. Which one of the follow-
ing is best to share with the care team regarding this
A. Administer 10 vials of digoxin immune Fab.
patient’s medications?
B. Administer 1 vial of digoxin immune Fab and
recheck digoxin concentration in 12 hours. A. Nifedipine can cause HF because it causes edema.
C. Hold digoxin until SCr normalizes and closely B. Metformin exacerbates HF because it causes lactic
monitor K. acid production in cardiac tissue.
D. Continue digoxin and obtain a serum potassium C. Lisinopril exacerbates HF because it decreases the
concentration every 4–6 hours. glomerular filtration rate and causes edema.
D. Clozapine can cause HF through IgE-mediated
25. A 61-year-old man has a medical history that includes
hypersensitivity reactions.
HF (EF 30%), atrial fibrillation, and depression. His home
drugs include furosemide 60 mg twice daily, lisinopril 10 28. A 40-year-old woman has a medical history that includes
mg daily, metoprolol succinate 100 mg daily, and apix- depression and atrial fibrillation with a recent community-
aban 5 mg twice daily. His atrial fibrillation symptoms acquired pneumonia. She is seen in the ED after experi-
continue to worsen despite optimization of β-blocker encing a syncopal episode while performing household
therapy. Which one of the following is best to recom- chores. The patient’s ECG reveals polymorphic ventric-
mend for this patient? ular tachycardia (VT) indicative of torsades de pointes
(TdP). Her home drugs include fluoxetine 20 mg daily,
A. Add dofetilide.
alprazolam 0.5 mg twice daily as needed, dofetilide 500
B. Change to metoprolol tartrate.
mcg twice daily, and clarithromycin 500 mg daily twice
C. Add diltiazem.
daily initiated 4 days ago. The cardiology team reviews
D. Add flecainide.
her medication profile; discontinues fluoxetine, clarithro-
26. A 65-year-old man presents to urgent care with fever, dys- mycin, and dofetilide; and corrects her hypokalemia. The
pnea, and a productive cough. Chest radiography reveals patient has a recurrence of TdP while in the ED but is
ground-glass opacities and new left lower lobe infiltrates. hemodynamically stable. Later that evening, after being
His WBC is elevated at 16.1 × 103 cells/mm3. An ECG reveals transferred to the ICU, she begins to experience nausea
normal sinus rhythm with a QTc of 465 milliseconds. and vomiting. In the setting of prolonged QTc, which one

PSAP 2022 Book 1 • Cardiology 54 Drug-Induced Cardiovascular Disease


of the following is the best antiemetic to recommend for A. Add amlodipine 10 mg daily.
this patient? B. Add hydrochlorothiazide 25 mg daily.
C. Discontinue tacrolimus.
A. Droperidol
D. Change tacrolimus to cyclosporine 150 mg twice
B. Ondansetron
daily.
C. Chlorpromazine
D. Promethazine 30. A 55-year-old man (weight 80 kg) presents to your primary
care clinic for a routine diabetes follow-up. His medical his-
29. A 62-year-old woman presents for an outpatient visit
tory includes HF with reduced EF and type 2 diabetes. His
with her cardiologist. Her medical history includes a kid-
home drugs include metformin 1000 mg twice daily, enal-
ney transplant 4 months ago, unstable angina, diabetes,
april 10 mg daily, carvedilol 12.5 mg twice daily, and furo-
and hypertension. Her home drugs include tacrolimus
semide 60 mg twice daily. The patient’s vital signs today
4 mg twice daily (concentrations within normal range),
in the clinic are blood pressure 123/67 mm Hg and heart
prednisone 5 mg daily, mycophenolate mofetil 1000 mg
rate 73 beats/minute. Fasting laboratory tests at this visit
twice daily, metformin 1000 mg twice daily, aspirin 81 mg
include glucose 175 mg/dL, A1C 8.3%, SCr 1.2 mg/dL, and
daily, rosuvastatin 20 mg daily, losartan 50 mg daily, and
K 4.2 mEq/mL. Given his level of glucose control, which
atenolol 50 mg daily. The patient reports doing well other
one of the following is best to recommend for this patient?
than having high blood pressure readings when check-
ing her blood pressure at home. Her primary care phy- A. Pioglitazone 15 mg oral daily
sician recently doubled the doses of both atenolol and B. Saxagliptin 5 mg oral daily
losartan. Her physical examination is notable for blood C. Dapagliflozin 10 mg oral daily
pressure 168/96 mm Hg and heart rate 62 beats/minute. D. Insulin glargine 10 units subcutaneously
After confirming your plan with the transplant nephrolo-
gist, which one of the following is best to recommend for
this patient?

PSAP 2022 Book 1 • Cardiology 55 Drug-Induced Cardiovascular Disease


Learner Chapter Evaluation: Drug-Induced Cardiovascular Disease

As you take the posttest for this chapter, also evaluate the 25. The learning assessment activities used in the chapter
material’s quality and usefulness, as well as the achievement were effective.
of learning objectives. Rate each item using this 5-point scale: 26. The chapter was effective overall.

• Strongly agree 27. The activity met the stated learning objectives.
• Agree 28. If any objectives were not met, please list them here.
• Neutral
• Disagree OTHER COMMENTS
• Strongly disagree
29. Please provide any specific comments related to any
16. The content of the chapter met my educational needs. perceptions of bias, promotion, or advertisement of
17. The content of the chapter satisfied my expectations. commercial products.
18. The author presented the chapter content effectively. 30. Please expand on any of your above responses, and/or
provide any additional comments regarding this chapter:
19. The content of the chapter was relevant to my practice
and presented at the appropriate depth and scope.
20. The content of the chapter was objective and balanced. Questions 31–33 apply to the entire learning module.

21. The content of the chapter is free of bias, promotion, and 31. How long did it take you to read the instructional materi-
advertisement of commercial products. als in this module?
22. The content of the chapter was useful to me. 32. How long did it take you to read and answer the assess-
23. The teaching and learning methods used in the chapter ment questions in this module?
were effective. 33. Please provide any additional comments you may have
24. The active learning methods used in the chapter were regarding this module:
effective.

PSAP 2022 Book 1 • Cardiology 56 Drug-Induced Cardiovascular Disease


Cardiology II
Cardiology II Panel

Series Editors: Antithrombotic Therapy in Cardiac


Cynthia A. Sanoski, Pharm.D., FCCP, BCPS Interventions
Department Chair Authors
Associate Professor of Pharmacy Practice
Stephanie Dwyer Kaluzna, Pharm.D., BCCP
Thomas Jefferson University
Philadelphia, Pennsylvania Clinical Assistant Professor
Department of Pharmacy Practice
Daniel M. Witt, Pharm.D., FCCP, BCPS
University of Illinois Chicago College of Pharmacy
Professor and Chair Cardiovascular Clinical Pharmacist
Department of Pharmacotherapy University of Illinois Hospital and Health Sciences System
Assistant Dean of Clinical Affairs Chicago, Illinois
University of Utah College of Pharmacy
Jaclynne Gowen, Pharm.D., BCCP
Salt Lake City, Utah
Clinical Ambulatory Pharmacist
Faculty Panel Chair: Department of Cardiology
Wentworth-Douglass Hospital
Craig J. Beavers, Pharm.D., FCCP, FACC, FAHA, BCCP,
Dover, New Hampshire
BCPS-AQ Cardiology, CACP
Director of Cardiovascular Services Reviewers
Baptist Health Paducah
Cardiovascular Clinical Pharmacist A. Josh Roberts, Pharm.D., BCCP,
BCPS-AQ Cardiology, AACC
Department of Pharmacy Services
UK Healthcare Pharmacist Specialist - Cardiology
Assistant Adjunct Professor PGY2 Cardiology Pharmacy Residency Program Director
Department of Pharmacy Practice and Science UC Davis Medical Center
University of Kentucky College of Pharmacy Sacramento, California
Paducah, Kentucky Associate Professor of Pharmacy (WOS)
UC San Francisco School of Pharmacy
San Francisco, California
Peripheral Arterial Disease Bethany A. Ford, Pharm.D., BCPS
Author Assistant Professor
Anastasia L. Armbruster, Pharm.D., FACC, BCPS, BCCP Department of Clinical and Administrative Sciences
Notre Dame of Maryland University School of Pharmacy
Associate Professor
Baltimore, Maryland
Department of Pharmacy Practice
St. Louis College of Pharmacy at UHSP Christine S. Ji, Pharm.D., BCPS, BCCP
St. Louis, Missouri Clinical Pharmacy Specialist Cardiology
Department of Pharmacy
Reviewers Massachusetts General Hospital
Toby Trujillo, Pharm.D., FCCP, FAHA, BCPS Boston, Massachusetts
Associate Professor
Department of Clinical Pharmacy
University of Colorado Skaggs School of Pharmacy
Aurora, Colorado
Brenda L. Pahl, Pharm.D., BCPS
Assistant Dean of Student Affairs and Admissions
Assistant Professor of Pharmacy Practice
Cedarville University School of Pharmacy
Cedarville, Ohio
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
Consultancies: Anastasia Armbruster (ACC/AHA Joint Committee Clinical Practice Guidelines); Jessica Bente (ACCP); Allison
Burnett: Consultancies (Anticoagulation Forum; National Certification Board for Anticoagulation Providers; Global Strategies);
Jessie L. Dunne (Pulmonary Hypertension Association); Nancy M. Nix (Pharmacosmos); Daniel M. Riche (Novo Nordisk; Merck;
AstraZeneca); Ralph J. Riello III (AstraZeneca; Johnson & Johnson); Sara Vazquez (Anticoagulation Forum);

Stock Ownership:

Royalties: Allison Burnett (Wolters-Kluwer); Daniel M. Riche (McGraw-Hill); Honoraria (Merck); Sara Vazquez (UpToDate)

Grants: Anastasia Armbruster (ACCP); Jessica Bente (New Jersey Society of Health-System Pharmacists); Gregory Castelli
(American Board of Family Medicine); Kevin Cowart (University of South Florida Taneja College of Pharmacy); Ralph J. Riello
III (AstraZeneca)

Honoraria: Anastasia Armbruster (AstraZeneca); John Bucheit (Association of Diabetes Care & Education Specialists); Nancy M.
Nix (Coherus; Pfizer; AstraZeneca); Ralph J. Riello III (Alexion; AstraZeneca; Janssen)

Other:

Nothing to disclose: Katherine Aymond, Eugene N. Bush, Nicholas W. Carris, Stephanie Dwyer Kaluzna, Lindsey Federle, Beth-
any A. Ford, Stormi E. Gale, Jaclynne R. Gowen, Glenn Herrington, Christine Ji, Amy L. Lehnert, Nilam R. Naik, Brenda Pahl, Joel
J. Peterson, A. Joshua Roberts, Kelly M. Rudd, Ashley Schenk, Susan M. Smith, Elisabeth M. Wang, Toby C. Trujillo, Barbara Wig-
gins, Leslie Wooten, Eman El Sayed Younis

ROLE OF BPS: The Board of Pharmacy Specialties (BPS) is an autonomous division of the American Pharmacists Association
(APhA). To maintain its strict, independent standards for certification, BPS does NOT endorse or provide review information,
preparatory courses, or study guides for Board Certification Examinations. The Board, through its specialty councils, is respon-
sible for specialty examination content, administration, scoring, and all other aspects of its certification programs. BPS is totally
separate and distinct from ACCP. PSAP has been approved by BPS for use in BCPS recertification. Information about the BPS
recertification process is available online.

Questions regarding BPS specialty recertification should be directed to:

Board of Pharmacy Specialties


2215 Constitution Avenue NW
Washington, DC 20037
(202) 429-7591
Continuing Pharmacy Education
and Recertification Instructions
Continuing Pharmacy Education (CPE) Credit: The American College of Clinical Pharmacy is accredited by the
Accreditation Council for Pharmacy Education (ACPE) as a provider of CPE.

PSAP Target Audience: The target audience for PSAP 2022 Book 1 (Cardiology) is pharmacotherapy specialists and advanced
level clinical pharmacists encountering diverse cardiovascular patient populations.

Available CPE credits: Purchasers who successfully complete all posttests for PSAP 2022 Book 1 (Cardiology) can earn 21.5 contact
hours of CPE credit. The universal activity numbers are as follows: Cardiology I – 0217-0000-22-002-H01-P, 5.0 contact hours;
Cardiology II – 0217-0000-22-003-H01-P, 6.0 contact hours; Cardiology III – 0217-0000-22-004-H01-P, 5.0 contact hours; and
Cardiology IV – 0217-0000-22-005-H01-P, 5.5 contact hours. You may complete one or all available modules for credit. Tests may
not be submitted more than one time.

TO EARN CPE CREDITS FROM THIS PSAP BOOK

Posttest access: Go to www.accp.com and sign in with your e-mail address and password. Technical support is available from 8 a.m.
to 5 p.m. (Central) weekdays by calling (913) 492-3311. PSAP products are listed under My Products on your My Account page.

BCPS test deadline: 11:59 p.m. (Central) on July 15, 2022.


ACPE test deadline: 11:59 p.m. (Central) on January 18, 2025.

BCPS Recertification Credit: To receive BCPS recertification CPE credit, a PSAP posttest must be submitted within
the 6-month period after the book’s release (see above). Only completed tests are eligible for credit; no partial or
incomplete tests will be processed. You may complete one or all available modules for credit. Tests may not be
submitted more than one time.

The passing point to earn BCPS recertification credit is based on an expert analysis of the assessment items in each posttest
module. Any posttest submitted before the BCPS test deadline that meets this passing point will earn BCPS recertification
credits. These credits will be assigned as of the date of test submission and reported within 48 hours to BPS. For statements of
recertification credit, visit www.bpsweb.org.

Questions regarding the number of hours required for BCPS recertification should be directed to BPS at (202) 429-7591 or
www.bpsweb.org. The ACCP Recertification Dashboard is a free online tool that can track recertification credits as they are
earned through ACCP and schedule new opportunities for credits from upcoming ACCP professional development programs.

ACPE CPE Credit: To receive ACPE CPE credit for a PSAP module, a posttest must be submitted within 3 years after the book’s
release (see above). Only completed tests are eligible for credit; no partial or incomplete tests will be processed. You may
complete one or all available modules for credit. Tests may not be submitted more than one time.

Any posttest submitted before the ACPE deadline that scores 50% or greater will be awarded the appropriate CPE. These
credits will be assigned as of the date of test submission and reported within 48 hours. For statements of CPE credit, visit
www.mycpemonitor.net.

Posttest answers: The explained answers—with rationale and supporting references—will be posted 1 week after the BCPS test
deadline and will be available to anyone who has either (1) submitted a posttest or (2) waived the right to receive credit from a
posttest (see below). Go to www.accp.com and sign in with your e-mail address and password. Click the PSAP book on your My
Account page and you will see a link to the explained answers.

Test Waivers: To access the explained answers without submitting a posttest, sign in to your My Account page, select the PSAP
book, and click on the waiver link for that module. By completing the waiver form for a module, you waive the opportunity to
receive CPE credit for that module. After you submit a waiver, you will see a link to the PDF file that contains the answers for the
module you waived. Answers will be available starting 1 week after the BCPS test deadline.
Peripheral Arterial Disease
By Anastasia L. Armbruster, Pharm.D., FACC, BCPS, BCCP

Reviewed by Toby Trujillo, Pharm.D., FCCP, FAHA, BCPS; and Brenda L. Pahl, Pharm.D., BCPS

LEARNING OBJECTIVES

1. Assess patients for risk of peripheral arterial disease (PAD) and determine eligibility for cardiovascular risk reduction
strategies.
2. Assess the role of antithrombotic therapy after revascularization for the management of PAD.
3. Evaluate the role of emerging therapies in the management of PAD.
4. Develop a treatment plan for patients with a diagnosis of PAD.
5. Design a treatment plan for patients who present with critical or acute limb ischemia.

INTRODUCTION
ABBREVIATIONS IN THIS CHAPTER
Peripheral arterial disease (PAD) is a manifestation of systemic ath-
ABI Ankle-brachial index
erosclerosis and is part of a disease spectrum that includes coro-
ACC American College of Cardiology
nary artery disease (CAD) and cerebrovascular disease. A variety of
ACEI Angiotensin-converting enzyme
inhibitor terms have been used to describe this condition, including peripheral
ADA American Diabetes Association vascular disease, peripheral artery disease, and lower extremity arterial
disease. Atherosclerosis can develop in arterial beds throughout the
AHA American Heart Association
body. This chapter focuses on management of lower extremity PAD.
ALI Acute limb ischemia
Narrowing of vessels decreases blood flow, and occlusion can occur
ARB Angiotensin-receptor blockers
secondary to plaque rupture and/or thrombosis of narrowed ves-
ASCVD Atherosclerotic cardiovascular
disease sels. The most common sites are the femoral and popliteal arteries
CAD Coronary artery disease (Gerhard-Herman 2017). This anatomy is outlined in Figure 1. As
CDT Catheter-directed thrombolysis plaque builds and arteries are narrowed, an oxygen supply/demand
CLI Critical limb ischemia mismatch occurs and results in ischemia and extremity pain, known
as intermittent claudication (IC). This condition can be described as
CV Cardiovascular
fatigue and cramping or pain in the lower extremities elicited by walk-
DAPT Dual antiplatelet therapy
ing and relieved by rest, typically within 10 minutes. More serious
DM Diabetes mellitus
complications may occur with the progression of PAD, including acute
ESC European Society of Cardiology
limb ischemia (ALI), which is defined as an acute (less than 2 weeks)
IC Intermittent claudication
hypoperfusion of the limb. Patients may report pain, pulselessness
MACE Major adverse cardiovascular
events in the affected limb, or cold extremities. Critical limb ischemia (CLI) is
MALE Major adverse limb events characterized by chronic symptoms (more than 2 weeks), including
pain at rest, nonhealing ulcers, or gangrene (Gerhard-Herman 2017)
MI Myocardial infarction
(see Figure 1).
PAD Peripheral arterial disease
SBP Systolic blood pressure
Paucity of Data
SGLT2 Sodium-glucose co-transporter-2
Peripheral arterial disease remains grossly under recognized and
Table of other common abbreviations. underdiagnosed, with much of the data regarding treatment deci-
sions derived from secondary outcomes or sub-analyses of larger
cardiovascular (CV) trials that focus to a greater degree on athero-
sclerotic cardiovascular disease (ASCVD) in other vascular beds.

PSAP 2022 Book 1 • Cardiology 63 Peripheral Arterial Disease


Figure 1. Major arteries serving the lower limb are shown in anterior and posterior views.
Reprinted with permission from Lumen. Anatomy and Physiology II. Module 4: The Cardiovascular System: Blood Vessels and
Circulation.

This focus has left large knowledge gaps regarding the patho-
physiology and treatment of PAD. Major society guidelines
BASELINE KNOWLEDGE STATEMENTS have conflicting information regarding screening recommen-
dations and use of pharmacotherapy. Moreover, the most
Readers of this chapter are presumed to be familiar
recent advances in medications have not been incorporated
with the following:
into a major cardiovascular guideline. This lack of clarity has
• General knowledge of the pathophysiology that likely impacted patient care.
leads to development of atherosclerotic cardiovas-
cular disease Updated American Heart Association (AHA)/American Col-
lege of Cardiology (ACC) clinical practice guidelines are esti-
• Current guideline recommendations for the man-
agement of hypertension, hyperlipidemia, and type mated to be released in the second quarter of 2022. A study
2 diabetes mellitus examining prescribing patterns within the U.S. Department of
• Drug knowledge of antithrombotic therapy used to Veterans Affairs demonstrated that, compared with patients
reduce the risk of cardiovascular events who have CAD, patients with PAD alone were less likely to
receive guideline-directed medical therapy, including statin
Table of common laboratory reference values therapy, antiplatelet therapy, and achievement of A1C less
than 7% (Hira 2016). Similarly, the PARTNERS trial demon-
ADDITIONAL READINGS strated an overall low awareness of PAD diagnosis among
primary care physicians, as well as low use of guideline-
The following free resources have additional back-
directed medical therapy (Hirsch 2001). Risk factor recogni-
ground information on this topic:
tion and management is crucial in preventing the progression
• AHA PAD Go-To-Guide for Healthcare Professionals of PAD and the need for amputation. While large gaps exist in
• Tobacco Cessation for Patients with Cardiovascu- patient care, pharmacists have an opportunity to participate
lar Disease
in appropriate screening, risk reduction measures, and treat-
ment of patients with PAD. Engagement in research focusing

PSAP 2022 Book 1 • Cardiology 64 Peripheral Arterial Disease


on the management of PAD as well as educational efforts with major amputation. Patients who receive a diagnostic
within the health care community to improve overall treat- angiogram are significantly less likely to undergo amputa-
ment of this patient population are also needed. tion. However, African American patients and those with lower
incomes were less likely to undergo diagnostic angiograms
Epidemiology and Risk Factors during index hospitalizations. Access to specialized care also
Peripheral arterial disease is a marker for systemic athero- plays a crucial role in avoiding amputation because patients
sclerotic disease; these patients are likely to have disease are more likely to receive alternate management strategies
in other vascular beds—coronary, carotid, renal arteries, and at centers with specially trained physicians available and
the abdominal aorta. The incidence of PAD is estimated to be higher patient volume (Henry 2011). Efforts should be made
10%–15% and increases with age (more than 20% in patients to ensure patients have equal access to adequate PAD screen-
older than 80 years), affecting 8–10 million Americans ing and work-up before considering major amputation.
(Shu 2018). Box 1 summarizes characteristics of patients
at increased risk of PAD. These risk factors have varying
DIAGNOSIS
degrees of impact on disease progression. Diabetic patients
have 2–4 times the risk of developing PAD, CAD, and ischemic Appropriate screening of patients for PAD is crucial because
stroke. Regarding racial disparities, the likelihood of PAD is an estimated 50% of patients are asymptomatic or at least
50% higher in African American patients compared with report no symptoms without intentional screening (Shu
white patients (Shu 2018). Cigarette smoking has a stronger 2018). Screening of high-risk patients is recommended by the
association with development of PAD compared with CAD 2016 AHA/ACC guidelines. Early identification is important
and stroke (symptomatic PAD: HR 4.13; CAD: HR 2.1; stroke: to reduce the development of complications and advanced
HR 1.8) (Ding 2019). The association of PAD with smoking disease, combined with crucial risk reduction strategies. For
is not only the most significant compared with other CV dis- patients considered to have a high risk of PAD, see Box 1.
eases, but PAD is also correlated with quantity of cigarettes
smoked—both with number of cigarettes smoked per day and History and Physical Examination
number of years smoked (Ding 2019). Patients should receive Initial assessment for PAD can be performed in any clinical
aggressive risk factor management to reduce the risk of CV setting. A detailed clinical history and basic physical exam-
events and improve functional status (see Box 1). ination are the first steps in establishing a diagnosis; however,
diagnostic tests are required for confirmation. Although most
Health Care Disparities clinicians associate IC with PAD, only 10% of patients report
Although overall diagnosis and management of PAD must be this classic symptom at the time of diagnosis, with up to 40%
improved at a national level, significant health care disparities not reporting any leg pain (McDermott 2001). Therefore, the
exist, particularly with respect to management of advanced presence of symptoms alone is inadequate to identify every-
stages of the disease. After adjusting for patient-related one who will benefit from risk-reducing interventions, high-
factors, African American, Native American, and Hispanic lighting the important role of screening. Similar to CAD, not
patients are more likely to undergo major amputation as a all patients present with classic symptoms; atypical symp-
management strategy for CLI. Socioeconomic factors such as toms exist for lower extremity ischemia as well. These symp-
lower income and non-private insurance are also associated toms may include other exertional symptoms or perceived
walking impairment. Patients should be asked about all
non-joint related limb pain to fully evaluate for potential PAD
Box 1. Patients at Increased Risk of PAD symptoms. It is important to remember symptoms and their
• Age < 50 yr, with DM and 1 additional risk factor for intensity may vary among patients and do not necessarily
atherosclerosis correlate with degree of disease. The Edinburgh Claudication
• Age ≥ 50– 64 yr, with risk factors for atherosclerosis (DM, Questionnaire is recommended by the 2018 European Society
smoking, hyperlipidemia, hypertension) or family history of Cardiology (ESC) guideline as a symptom screening tool to
of PAD
use with patients and has been validated in several studies
• Age ≥ 65 yr (Leng 1992). Both the Fontaine and Rutherford classification
• Individuals with known atherosclerosis in another vascular
bed (e.g., coronary, carotid) systems can be used to categorize a patient’s description of
symptoms (Aboyans 2018; Gerhard-Herman 2017).
DM = diabetes mellitus; PAD = peripheral arterial disease. Physical examination is also crucial when screening for
Information from: Gerhard-Herman MD, Gornik HL, Barrett C, PAD. High-risk patients should undergo palpation of lower
et al. 2016 AHA/ACC guideline on the management of patients
with lower extremity peripheral artery disease: a report of the extremity pulses, auscultation for femoral bruits, and visual
American College of Cardiology/American Heart Association inspection of the legs and feet. Pulses can be described in
Task Force on Clinical Practice Guidelines. J Am Coll Cardiol
2017;69:e71-126.
the following manner: absent (0), diminished (1), normal (2),
or bounding (3). An absent or diminished posterior tibial

PSAP 2022 Book 1 • Cardiology 65 Peripheral Arterial Disease


Table 1. Resting ABI Interpretation in Symptomatic Patients

Resting ABI Interpretation Recommendation

≤0.90 Abnormal Diagnostic for PAD


0.91–0.99 Borderline Exercise ABI necessary to establish diagnosis of PAD
1.00–1.40 Normal Exercise ABI necessary to establish diagnosis of PAD
>1.40 Noncompressible Perform TBI; ≤0.7 is diagnostic of PAD

ABI = ankle-brachial index; TBI = toe-brachial index; PAD = peripheral arterial disease.
Information from: Gerhard-Herman MD, Gornik HL, Barrett C, et al. 2016 AHA/ACC guideline on the management of patients with
lower extremity peripheral artery disease: a report of the American College of Cardiology/American Heart Association Task Force
on Clinical Practice Guidelines. J Am Coll Cardiol 2017;69:e71-126.

pulse, compared with dorsalis pedis pulse, is most consis- slope; then the test is stopped when the patient can walk
tent with PAD. Several abnormal physical findings increase no further because of pain. A post-exercise ABI decrease of
the likelihood of a confirmatory PAD diagnosis, whereas a more than 20% is diagnostic for PAD (Aboyans 2018). Alter-
normal examination absent of bruits decreases the likelihood native tests are available if a treadmill is unavailable. The
of PAD. Other potential physical findings include limb hair 6-minute walk test has previously been correlated with the
loss, shiny skin, or muscle atrophy. Physical findings such as hemodynamic severity of PAD (Montgomery 1998). The pedal
cool extremities or non-healing ulcers are more consistent plantarflexion ABI test has also been suggested as an alter-
with advanced disease, including CLI or ALI (Aboyans 2018; native to treadmill testing (Gerhard-Herman 2017).
Gerhard-Herman 2017).
Diagnostic Imaging
Diagnostic Testing Additional testing, including diagnostic imaging, is reserved
Resting Ankle-Brachial Index for patients undergoing revascularization. These tests
Ankle-brachial index (ABI) should be the first diagnostic test may include duplex ultrasonography, CT angiography, or
after a thorough history and clinical examination. An ABI less MRI angiography. Selection of specific diagnostic testing
than 0.9 is both sensitive and specific for diagnosis. Sensi- should consider patient-specific factors, including risk of
tivity can be decreased by diabetes mellitus (DM) and end- contrast-induced nephropathy. It is important to note that
stage chronic kidney disease (Aboyans 2012). A resting ABI is diagnostic angiography should not be performed in asymp-
performed with the patient in the supine position for at least tomatic patients because of a lack of benefit and risk of harm
10 minutes while brachial blood pressure is checked in both (Gerhard-Herman 2017).
arms. The highest systolic blood pressure (SBP) should be
used to perform the ABI calculation for each leg. Pressures PHARMACOTHERAPY IN PATIENTS
should then be obtained using a continuous-wave Doppler WITH PAD
device on the dorsalis pedis and posterior tibial arteries of The mainstay of treatment in patients with PAD is addressing
the legs. The ABI is the ratio of the highest pressure in each modifiable risk factors with the goals of reducing CV risk and
leg and the obtained SBP. Table 1 outlines appropriate inter- disease progression, including major amputation. There is a
pretation and follow-up testing of ABI results in symptom- lack of high-quality, randomized controlled trials evaluating
atic patients. Up to 30% of symptomatic patients with normal pharmacotherapy specifically in patients with PAD. Currently,
resting ABI studies may have an abnormal ABI after exercise the strongest recommendations exist for antiplatelet and sta-
(Stein 2006). Exercise ABI testing is also indicated in patients tin therapies, as discussed in following text. Table 2 outlines
with borderline results. Of note, ABI can be falsely elevated in the pharmacotherapy recommendations from both the 2016
patients with advanced diseased related to severely calcified, AHA/ACC and 2018 ESC guidelines.
noncompressible arteries.
Hypertension
Exercise ABI Current AHA/ACC guidelines recommend that patients
A treadmill test can be performed for functional assessment receive antihypertensive therapy to reduce the risk of myo-
and in symptomatic patients with normal resting ABIs. No cardial infarction (MI), stroke, heart failure, and CV death.
specific treadmill protocol is recommended over another. Although a specific blood pressure goal is not provided, cli-
The ESC guidelines recommend a speed of 3 km/hour at 10% nicians should follow the current guidelines for treating

PSAP 2022 Book 1 • Cardiology 66 Peripheral Arterial Disease


Table 2. ACC/AHA and ESC Recommendations for Medical Management of PAD

Recommendations ACC/AHA ESC


COR, LOE Class, LOE

Antiplatelet Therapy

Antiplatelet monotherapy (either aspirin 75–325 mg/day or clopidogrel 75 mg/day) to IA IA


reduce myocardial infarction, stroke and vascular death in symptomatic patients

Clopidogrel may be preferred to aspirin in patients who require antiplatelet therapy — IIb, B

Antiplatelet monotherapy is reasonable to reduce the risk of MACE in asymptomatic IIa, C-EO III, A
patients with ABI ≤ 0.90

Efficacy of dual antiplatelet therapy with aspirin and clopidogrel to reduce the risk of IIb, B-R —
MACE in symptomatic patients is not well established

Overall clinical benefit of adding vorapaxar to aspirin or clopidogrel in symptomatic IIb, B-R —
patients is uncertain

DAPT may be reasonable to reduce the risk of limb-related events in symptomatic IIb, C-LD IIb, B/C
patients after lower limb revascularization

Anticoagulation

Vitamin K antagonists should not be used to reduce the risk of CV ischemic events in III: Harm, A —
patients

Statin Therapy

Treatment with a statin medication is recommended for all patients I, A I, A

Reduction of LDL cholesterol to < 70 mg/dL or decrease by >50% is recommended if — I, C


baseline values are 70–135 mg/dL

Antihypertensive Therapy

Angiotensin-converting enzyme inhibitors or angiotensin-receptor blockers can be IIa, A IIa, B


effective to reduce the risk of CV ischemic events in hypertensive patients

Smoking Cessation

Smoking cessation is recommended in all patients I, A I, B

Patients who smoke cigarettes should be helped to develop a plan for quitting that I, A —
includes pharmacotherapy (i.e., varenicline, bupropion, and/or nicotine replacement
therapy) and/or referral to a smoking cessation program

Glycemic Control

In diabetic patients, glycemic control is recommended and should be coordinated I, C-EO I, C


between health care team members

Symptomatic Treatment of Claudication

Cilostazol is an effective therapy to improve symptoms and increase walking distance I, A —


in patients with claudication

Pentoxifylline is not effective for treatment of claudication III: No benefit, —


B-R

ABI = ankle-brachial index; ACC = American College of Cardiology; AHA = American Heart Association; C = consensus; COR = class of
recommendation; CV = cardiovascular; DAPT = dual antiplatelet therapy; EO = expert opinion; ESC = European Society of Cardiology;
LD = limited data; LOE = level of evidence; MACE = major adverse cardiac event; PAD = peripheral artery disease; R = randomized.
Information from: Aboyans V, Ricco J, Bartelink M, et al. 2017 ESC guidelines on the diagnosis and treatment of peripheral arterial
diseases, in collaboration with the European Society for Vascular Surgery. Eur Heart J 2018;39:763-816; Gerhard-Herman MD,
Gornik HL, Barrett C, et al. 2016 AHA/ACC guideline on the management of patients with lower extremity peripheral artery disease:
a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll
Cardiol 2017;69:e71-126.

PSAP 2022 Book 1 • Cardiology 67 Peripheral Arterial Disease


hypertension (Gerhard-Herman 2017). Therefore, patients
Box 2. High-Risk Conditions for
with PAD should target a blood pressure of 130/80 mm Hg or
Developing Peripheral Arterial Disease
less, consistent with the 2017 ACC/AHA guideline for manag-
ing blood pressure (Whelton 2018). A J-shaped relationship • Age ≥ 65 yr
has been noted among patients with PAD regarding SBP and
• Chronic kidney disease (estimated glomerular filtration rate
15–59 mL/min/1.73 m2)
occurrence of CV events in a post hoc analysis of the INVEST • Current smoking
trial. The composite outcome of all-cause death, non-fatal • Diabetes mellitus
MI or stroke occurred least often with an average treated • Heterozygous familial hypercholesterolemia
SBP of 135–145 mm Hg. For patients who achieving SBP of • History of congestive heart failure
110 mm Hg or less, the occurrence of the primary outcome • History of coronary artery bypass graft surgery or percuta-
neous coronary intervention other than for management of
increased compared with patients without PAD (HR 1.69 vs. a major atherosclerotic cardiovascular disease event
0.99; p=0.04) (Bavry 2010). • Hypertension
The use of angiotensin-converting enzyme inhibitors • Persistently elevated LDL cholesterol (≥100 mg/dL) despite
(ACEIs) or angiotensin-receptor blockers (ARBs) is reason- maximally tolerated statin therapy and ezetimibe
able in patients with PAD (class of recommendation IIa, A)
Information from: Grundy SM, Stone NJ, Bailey AL, et al. 2018
(Gerhard-Herman 2017). A subgroup analysis of the HOPE AHA/ACC/AACVPR/AAPA/ABC/ACPM/ ADA/AGS/ APhA/ASPC/
trial evaluated patients who were normotensive randomized NLA/PCNA guideline on the management of blood cholesterol:
a report of the American College of Cardiology/American Heart
to ramipril or placebo and followed for 4.5 years. Patients Association Task Force on Clinical Practice Guidelines. J Am
with an ABI of 0.90 of less were enrolled, with or without Coll Cardiol 2019;73:e285-350.
symptoms. In the subgroup of 4051 patients with PAD, rami-
pril reduced the risk of MI, stroke, or vascular death by 25%
(Ostergren 2004). Similar results were noted in the ONTARGET than 70 mg/dL. For the highest risk patients, addition of a
trial, which compared telmisartan versus ramipril, thus sup- proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibi-
porting the use of ARBs as an alternative agent (ONTARGET tor should be considered. Highest risk patients include those
Investigators 2008). The ESC guidelines also list calcium with symptomatic PAD (i.e., ABI less than 0.85 with history
channel blockers as preferred agents based on their poten- of claudication, or previous revascularization or amputation)
tial for peripheral arterial dilation. In addition, some data sug- and one other ASCVD event or several high-risk conditions,
gest β-blockers with vasodilation properties may also offer as defined in the guidelines (Grundy 2019). High-risk con-
benefit related to increased walking distance. Nebivolol ditions are outlined in Box 2. These recommendations are
demonstrated a 31% increase in walking distance in patients based on both the IMPROVE-IT trial and a sub-analysis of the
with claudication and hypertension. Based on the additional FOURIER trial among patients with a history of MI, stroke, or
nitric-oxide releasing properties, nebivolol may have benefit PAD. These study results showed that evolocumab reduced
in PAD compared with other β-blockers (Espinola-Klein 2011, the risk of major adverse limb events (MALE), including ALI,
Maffei 2009). Other than ACEIs/ARBs, no CV outcome data major amputation, and urgent revascularization (HR 0.58; 95%
are available to guide the selection of antihypertensive ther- CI, 0.38–0.88) (Bonaca 2018; Cannon 2015). Data exists for
apy. Patients with PAD often have other comorbid disease the PCSK9 inhibitor, alirocumab as well, from a pre-specified
states, which should be considered when selecting antihy- sub-analysis of the ODYSSEY OUTCOMES trial. Authors
pertensive pharmacotherapy. noted the absolute risk reduction increased in patients who
had additional types of cerebrovascular disease, demonstrat-
Dyslipidemia ing that patients with PAD had an added benefit with PCSK9
Statin therapy is the cornerstone of lipid management in inhibitors, although patients with PAD were a small percent-
patients with PAD. Data from the REACH registry demon- age of the trial population (Jukema 2019). It is important to
strated a reduction in adverse limb outcomes, including wors- follow guideline recommendations to reach a target of 70
ening claudication, new episode of CLI, new percutaneous/ mg/dL or less in patients with known PAD, this may require
surgical revascularization, or amputation, in statin users ver- the addition of ezetimibe and/or a PCSK9 inhibitor. The 2018
sus non-users [22.0% vs. 26.2%; HR 0.82; 95% CI, 0.72–0.92). AHA/ACC guideline on the management of blood cholesterol
A statistically significant reduction in the composite of CV should also be followed for patients at high-risk of develop-
death, MI, and stroke was also noted (Kumbhani 2014). The ing PAD, similar to any patient at risk of ASCVD (Grundy 2019)
2018 AHA/ACC guideline on the management of blood choles- (see Box 2).
terol includes PAD as part of clinical ASCVD, which provides
additional guidance on lipid management. Patients should Diabetes Mellitus
receive a high-intensity statin to achieve a 50% or greater The presence of DM increases the risk of adverse PAD out-
reduction in LDL cholesterol, and they may be considered comes, including progression to CLI, amputation, and mor-
for addition of ezetimibe if LDL cholesterol remains greater tality. Diabetes is especially important to address because

PSAP 2022 Book 1 • Cardiology 68 Peripheral Arterial Disease


nontraumatic lower extremity amputation increased by 50% stroke, and PAD. A stronger association with smoking dura-
in patients with DM between 2009 and 2015 (Virani 2020; tion and intensity (patient-reported pack-years) was consis-
Gerhard-Herman 2017). Similar to other pharmacotherapy, tently observed with PAD compared with stroke and CAD.
specific data are limited regarding which anti-hyperglycemic Current smoking of 1 pack or more per day had the high-
agents have benefit in patients with PAD. Current American est association with PAD (HR 5.36; 95% CI, 4.16–6.91) com-
Diabetes Association (ADA) standards recommend priori- pared with CAD (HR 2.38; 95% CI, 2.08–2.73) and stroke (HR
tizing agents associated with reduction in CV death. These 1.88; 95% CI, 1.57–2.26). Smoking cessation was associated
agents include either a glucagon-like peptide-1 (GLP-1) recep- with the greatest risk reduction for PAD among the three ath-
tor agonist (i.e., liraglutide, semaglutide, dulaglutide) or a erosclerotic diseases (Ding 2019). This finding dramatically
sodium-glucose co-transporter-2 (SGLT2) inhibitor (i.e., cana- highlights the need for smoking cessation for both patients
gliflozin, dapagliflozin empagliflozin). Both should be consid- at risk of PAD and those with a diagnosis of PAD to prevent
ered regardless of A1C target. Figure 9.1 in the ADA standards disease progression. This point was clearly demonstrated
outlines current treatment recommendations (ADA 2021a). with 5-year outcomes in an observational cohort study of
Previously, a boxed warning was issued for canagliflozin 739 patients with advanced PAD who underwent angiog-
related to an increased risk of amputation. This warning was raphy for CLI or claudication. Patients who successfully
primarily based on the CANVAS trial results, in which the risk quit smoking had a decreased rate of all-cause mortality
of amputations was increased in patients receiving canagli- (14% vs. 31%; HR 0.33; 95% CI, 0.13–0.80) and improved
flozin versus placebo (6.3 vs. 3.4 per 1000 patient-years; HR amputation-free survival (81% vs. 60%; adjusted HR 0.40;
1.97; 95% CI, 1.41–2.75) (Neal 2017). Since that time, addi- 95% CI, 0.19–0.83) compared with patients who continued to
tional benefits have been demonstrated and lower rates of smoke (Armstrong 2014). Smoking cessation is difficult to
amputation with canagliflozin have been reported. The risk achieve, and patients often require several attempts to quit.
of amputation has also not been statistically significant in Clinicians may lack comfort in prescribing pharmacotherapy
other landmark SGLT-2 inhibitor trials. For these reasons, the in patients with cerebrovascular disease, which is specifi-
FDA removed the boxed warning regarding an increased risk cally addressed in the 2018 ACC Expert Consensus Decision
of amputations. A subgroup analysis of EMP-REG OUTCOME, Pathway on Tobacco Cessation Treatment (Barua 2018). Cur-
a landmark trial for empagliflozin, demonstrated consistent rent AHA/ACC guidelines provide a class of recommendation
CV benefit in patients with DM and PAD without an increased I level of evidence A recommendation for current smokers to
risk of amputation (5.5% vs. 6.3%; HR 0.84; 95% CI, 0.54–1.32) receive assistance in developing a plan to quit that includes
(Verma 2018). One meta-analysis compared the association pharmacotherapy with varenicline, bupropion, and/or nico-
of amputation in patients with PAD between SGLT-2 inhib- tine replacement therapy (Gerhard-Herman 2017).
itors and dipeptidyl peptidase-4 inhibitors (DPP4Is). Over-
all lower rates of heart failure and adverse lower limb events Antiplatelet Therapy
were reported for patients with PAD receiving SGLT-2 inhib- Antiplatelet agents are the cornerstone of therapy in patients
itors, compared with patients receiving DPP4Is (Lee 2020). with PAD to prevent limb events, MI, stroke, and CV death. Lit-
This evidence mitigates previous concerns related to ampu- erature is limited regarding an optimal antiplatelet strategy,
tations in patients receiving SGLT-2 inhibitors (Lee 2020). including agents and duration, and is derived from subgroup
Because PAD is under the ASCVD umbrella, the ADA stan- analyses of trials to evaluate primarily CAD. Current AHA/ACC
dards of care are an excellent resource regarding DM man- guidelines recommend monotherapy with aspirin (75–325
agement and appropriate agent selection. Like hypertension, mg/day) or clopidogrel (75 mg/day) to reduce the risk of MI,
it is always important to consider patient-specific factors and stroke, and vascular death in patients with symptomatic PAD
comorbid conditions when selecting glucose-lowering ther- (Gerhard-Herman 2017).
apy (ADA 2021a).
Aspirin
Smoking Cessation The strongest data in support of aspirin monotherapy to
Smoking is one of the most important modifiable risk fac- reduce major adverse cardiovascular events (MACE) are from
tors to prevent progression of PAD. The risks of MI, death a meta-analysis from the Antithrombotic Trialists’ Collabora-
and amputation are substantially higher in patients with tion (ATC). A 23% reduction in the odds of MACE was observed
PAD who continue to smoke compared with those who stop in 9214 patients with PAD across 42 trials (p=0.004) (ATC
(Armstrong 2014). The AHA/ACC guidelines currently recom- 2002). Aspirin has clear benefit in patients with symptomatic
mend counselling on smoking cessation at every office visit PAD, whereas the benefits of use are less clear in patients
(Gerhard-Herman 2017). A recent study using data from the who remain asymptomatic. In a randomized controlled trial of
Atherosclerosis Risk in Communities (ARIC) database eval- patients followed for 8.2 years who were screened, had an ABI
uated the association between smoking cessation and inci- less than 0.95, and were without symptoms, no statistically
dence of the three major atherosclerotic diseases—CAD, significant difference was evident for patients who received

PSAP 2022 Book 1 • Cardiology 69 Peripheral Arterial Disease


aspirin (13.7 events per 1000 person-years vs. 13.3; HR 1.03; with ticagrelor 60 mg twice daily combined with aspirin 81
95% CI, 0.84–1.27) (Fowkes 2010). Although available data do mg/day in stable patients with history of MI. Patients in this
not support the efficacy of aspirin in patients with asymptom- trial could also be randomized to ticagrelor 90 mg twice daily.
atic PAD, it is important to consider the increased risk of other A sub-analysis evaluating patients in the placebo arm (aspirin
CV events, especially when disease is present in another vas- only) with confirmed PAD at enrollment (5%) had more than a
cular bed. 2-fold increase in MACE versus patients without PAD (19.3%
vs. 8.4%; unadjusted HR 2.46; 95% CI, 1.92–3.15). An increase
Single Antiplatelet Therapy with P2Y12 in bleeding was observed compared with aspirin alone.
Inhibitors Patients with PAD and a history of MI appeared to derive
Both clopidogrel and ticagrelor have been studied as single even more benefit from dual antiplatelet therapy (DAPT) than
antiplatelet agents for treatment of symptomatic PAD. The patients with only CAD (Bonaca 2016). Currently, AHA/ACC
CAPRIE trial assessed 19,185 patients and compared aspirin guidelines highlight that the effectiveness of DAPT in symp-
325 mg/day with clopidogrel 75 mg/day in those with ath- tomatic patients is not well established (class IIb recommen-
erosclerotic vascular disease, which included a subgroup of dation) (Gerhard-Herman 2017).
patients with symptomatic PAD. Patients were followed for
1.9 years with a primary outcome of reduction in MACE. The Vorapaxar
PAD subgroup analysis of 6452 patients showed that clopi- Vorapaxar is a novel antagonist of protease-activated
dogrel was associated with fewer ischemic events than aspi- receptor-1, the primary platelet receptor for thrombin. This
rin (3.71% vs. 4.86%; HR 0.76; 95% CI, 0.64–0.91) (CAPRIE drug received FDA approval in 2014 for the reduction of
Steering Committee 1996). Overall differences in bleeding thrombotic CV events in patients with history of MI or PAD
rates between groups were not statistically significant; how- in combination with aspirin and/or clopidogrel. The TRA 2oP-
ever, GI bleeding was more common in patients treated with TIMI 50 trial was a randomized, double-blind, placebo-control
aspirin compared with clopidogrel (2.66% vs. 1.99%, p<0.05) trial that enrolled 26,449 patients with a history of MI, stroke,
(CAPRIE Steering Committee 1996). Based on these data, or symptomatic PAD. Of those enrolled, 5845 patients had
ESC guidelines prefer clopidogrel over aspirin in patients known PAD at the start of the trial, including those with
with PAD. polyvascular disease (Morrow 2012). In this sub-analysis,
A large randomized controlled trial in patients with symp- vorapaxar did not reduce the rates of MACE compared with
tomatic PAD compared monotherapy with ticagrelor 90 mg placebo in patients with PAD (11.9% vs. 11.3%; HR 0.94; 95%
twice daily to clopidogrel 75 mg/day. The EUCLID trial included CI, 0.78–1.14). In addition, rates of GUSTO (Global Use of
13,885 patients with symptomatic PAD, defined as an ABI less Strategies to Open Occluded Coronary Arteries) moderate or
than 0.80 or prior revascularization of the lower limbs, with a severe bleeding were increased (7.4% vs. 4.5%; HR 1.62; 95%
primary efficacy end point of MACE within 30 months. Tica- CI, 1.21–2.18), regardless of concomitant antiplatelet therapy
grelor was not superior to clopidogrel at reducing the inci- (Bonaca 2013). It is important to note vorapaxar is contrain-
dence of MACE (10.8% vs. 10.6%; HR 1.02; 95% CI, 0.92–1.13). dicated in patients with history of stroke, transient ischemic
No difference in major bleeding was noted (1.6%vs. 1.6%; HR attack, or intracerebral hemorrhage. Based on the available
1.10; 95% CI, 0.84–1.43) (Hiatt 2017). Patients also discontin- literature, current AHA/ACC guidelines describe the overall
ued ticagrelor more often due to dyspnea and minor bleed- clinical benefit of vorapaxar to be uncertain in patients with
ing. Ticagrelor could present limitations related to cost and symptomatic PAD (Gerhard-Herman 2017).
adverse effects, primarily dyspnea, however; this drug could
be considered in patients with concomitant acute coronary Oral Anticoagulation
syndrome and PAD. In addition to antiplatelet therapy, a growing body of litera-
ture is addressing the use of oral anticoagulants in patients
Dual Antiplatelet Therapy with PAD. Historically, anticoagulation has not been recom-
Dual aspirin and clopidogrel therapy versus aspirin alone was mended to reduce the risk of CV events in this patient pop-
studied in the CHARISMA trial, which included patients with ulation. In the WAVE trial, warfarin was combined with an
established CV disease (including symptomatic PAD) or sev- antiplatelet agent, primarily aspirin, in patients with PAD.
eral atherothrombotic risk factors. There was no difference Combination therapy with warfarin was not more effective
in MACE observed in the general trial population. However, in and was associated with an increase in life-threatening bleed-
a post hoc analysis of those with polyvascular disease (CAD ing compared with antiplatelet therapy alone (Anand 2007).
plus PAD) there was an increased risk of MACE and acute Edoxaban, a direct factor Xa inhibitor, has been inves-
limb events. A reduction in MI and ischemic hospitalization tigated in patients with recent endovascular treatment
was observed, with an increased in minor bleeding in those for PAD management. The ePAD study was a randomized,
patients receiving DAPT (Cacoub 2009). The PEGASUS-TIMI open-label trial that evaluated the combination of edoxaban
54 trial demonstrated a reduction in CV death, MI, or stroke 60 mg/day plus aspirin 100 mg/day or a clopidogrel 300-mg

PSAP 2022 Book 1 • Cardiology 70 Peripheral Arterial Disease


loading dose, followed by edoxaban 75 mg/day plus aspi- no increase in Thrombolysis in Myocardial Infarction major
rin 100 mg/day for 3 months after femoropopliteal interven- bleeding. A significant increase in ISTH major bleeding was
tion. The study assessed 203 patients who were followed for observed in the rivaroxaban plus aspirin group (5.94% vs.
6 months. For the primary efficacy end point of restenosis, 4.06%; HR 1.42; 95% CI, 1.10–1.84), but no increase in intra-
no difference was found between the two groups (30.9% vs. cranial hemorrhage or fatal bleeding was noted (Bonaca
34.7%; RR 0.89; 95% CI, 0.59–1.34; p=0.643), and no differ- 2020). Concomitant clopidogrel was explored in more detail
ence in International Society on Thrombosis and Hemosta- in a subgroup analysis. Rivaroxaban demonstrated a consis-
sis (ISTH) or Thrombolysis in Myocardial Infarction bleeding tent reduction in the primary outcome regardless of clopido-
score was found. Although this study did not find a differ- grel use (HR 0.85; 95% CI, 0.71–1.01 with clopidogrel vs. HR
ence, it was likely underpowered and concern was cited for 0.86; 95% CI, 0.73–1.01 without clopidogrel). Rivaroxaban was
higher risk of restenosis because of comorbid conditions in associated with more ISTH major bleeding within 365 days
the edoxaban group (Moll 2018). It is important to note within (HR 3.20; 95% CI, 1.44–7.13), compared with shorter durations
6 months there was a 30% risk of restenosis in both groups, of clopidogrel. Based on this analysis, it should be noted that
which highlights the difficulty of managing this patient triple therapy should be used with caution and limited to 30
population. days (Hiatt 2020). Based on these landmark trials, rivarox-
Rivaroxaban, a direct factor Xa inhibitor, has been inves- aban is now indicated in combination with aspirin to reduce
tigated in patients with PAD in two large randomized clinical the risk of major thrombotic vascular events in patients with
trials. The COMPASS study was a randomized, double-blind, PAD, specifically including those who have recently under-
placebo-control trial in stable CV disease, including patients gone a revascularization procedure.
with both CAD and PAD, which enrolled almost 27,000 For patients with favorable bleeding risk and symptom-
patients. Patients received rivaroxaban 2.5 mg twice daily atic PAD or with recent revascularization, it is reasonable to
(defined as low-dose) plus aspirin 100 mg, rivaroxaban 5 replace aspirin alone with low-dose rivaroxaban and aspirin. It
mg twice daily, or aspirin 100 mg/day. The trial was stopped is also important to consider a patient’s ability to afford rivar-
early because of significant reduction in the primary end oxaban; clopidogrel is generic and in many clinical scenar-
point in the treatment group (4.1% vs. 5.4%; HR 0.76; 95% CI, ios may provide a comparable alternative. Patients receiving
0.66–0.86); however, more major bleeding events occurred hemodialysis were excluded from both trials, and no dosing
in patients in the rivaroxaban plus aspirin group (3.1% vs. recommendations currently exist for this population. At this
1.9% HR 1.70; 95% CI, 1.40–2.05). No difference was noted in time, rivaroxaban is the only oral anticoagulant with compel-
fatal or intracranial bleeding, and most of the excess bleed- ling data in this patient population. The AGRIPPA trial will
ing was attributed to the GI tract (Eikelboom 2017). The PAD investigate the efficacy and safety of apixaban 2.5 mg twice
subgroup analysis included 6391 patients who could have daily plus aspirin compared with clopidogrel plus aspirin
history of revascularization, symptoms of PAD with objec- in patients with CLI undergoing endovascular intervention
tive confirmatory testing, or presence of CAD with ABI less (clinicaltrials.gov/NCT04229264).
than 0.90. Use of rivaroxaban 2.5 mg twice daily plus aspi-
rin 100 mg/day was associated with a reduction in MACE,
SYMPTOMATIC MANAGEMENT
as well as incidence of MALE compared with aspirin alone
in this population. Similar to the primary analysis, rivarox- Cilostazol
aban plus aspirin resulted in an increase in major bleeding Cilostazol, a phosphodiesterase 3 inhibitor, provides poten-
compared with aspirin alone (Anand 2018). Of note, the most tial benefit to patients with PAD through reversible inhi-
recent ADA guidelines state that combination therapy with bition of platelet aggregation and vasodilation. Current
aspirin and low-dose rivaroxaban should be considered in AHA/ACC guidelines note that cilostazol is an effective ther-
patients with stable CAD and/or PAD with a low bleeding risk apy to improve symptoms and increase walking distance in
(ADA 2021b). patients with IC (Gerhard-Herman 2017). A 2014 Cochrane
Most recently, the VOYAGER PAD trial evaluated 6564 review evaluating cilostazol included 15 randomized con-
patients with PAD undergoing revascularization, comparing trolled trials with 3718 patients. Cilostazol doses ranged from
rivaroxaban 2.5 mg twice daily plus aspirin 100 mg/day to aspi- 50–150 mg twice daily and were compared with both pent-
rin 100 mg/day alone. Clopidogrel was allowed at the discre- oxifylline and placebo. The pooled results demonstrated
tion of investigator for up to 6 months. Of those randomized, improved initial claudication distance and absolute claudi-
50.6% received clopidogrel for a median of 29 days. Patients cation distance with cilostazol. Few studies reported MI and
were randomized based on revascularization approach (sur- stroke, precluding any conclusions regarding the impact of
gical, 35%; endovascular, 66%) and planned use of clopidogrel cilostazol on these outcomes. Some adverse effects occurred
(50% in each arm). The combination of rivaroxaban and aspi- more often with cilostazol, including headache, diarrhea, diz-
rin was associated with a reduction in a composite of MALE ziness, and palpitations (Bedenis 2014). In an update to the
and MACE (17.3% vs. 19.9%; HR 0.85; 95% CI, 0.76–0.96), with previous Cochrane review, in which a pooled analysis was

PSAP 2022 Book 1 • Cardiology 71 Peripheral Arterial Disease


not performed because of differences in measures used Patients with CLI present with chronic (more than 2 weeks)
and reporting, limited data indicated no difference between symptoms, such as pain at rest or ulceration. Revasculariza-
cilostazol and pentoxifylline (Brown 2021). Cilostazol is con- tion is a priority to improve arterial blood flow. Selection of
traindicated in patients with heart failure at any stage because revascularization strategy depends on patient-specific fac-
of an observed decrease in survival for patients with stage tors and the expertise of available providers. Patients should
III and IV heart failure. Although cilostazol is recommended receive imaging before a decision regarding revascularization
by the 2016 AHA/ACC guidelines, 2017 ESC guidelines do not strategy. A key factor in decision-making for patients with
make a formal recommendation and call into question clinical CLI is the lesion characteristics—location, length, and cal-
usefulness of the medication (Aboyans 2018; Gerhard-Her- cification. An observational study of Medicare beneficiaries
man 2017). reported 4-year mortality rate from 49.3%–54.7% and major
amputation rates from 7.8%–10.8%. Although these findings
Pentoxifylline were statistically significant because of the large sample
Pentoxifylline, a blood viscosity reducer, is approved size, the authors questioned the clinical significance because
for the symptomatic management of IC. A randomized, of the observational study design. All treatment groups (per-
double-blind, placebo-controlled trial compared cilostazol cutaneous angioplasty, stent placement, atherectomy, or
100 mg twice daily to pentoxifylline 400 mg three times daily surgical bypass) were numerically similar, with very high mor-
or placebo. The primary outcome, maximal walking distance, tality rates regardless of revascularization approach (Musta-
was assessed every 4 weeks for 24 weeks. Mean maximal pha 2019).
walking distance for those receiving cilostazol increased by
54% compared to pentoxifylline and placebo. Improvement Endovascular Revascularization
in the pentoxifylline group was similar to placebo (Dawson Use of endovascular therapy for CLI management contin-
2000). Current AHA/ACC guidelines do not recommend the ues to increase. From 2003 to 2011, the use of endovascu-
use of pentoxifylline due to lack of benefit (Gerhard-Herman lar procedures significantly increased (5.1%–11.0%) and was
2017). associated with lower rates of in-hospital mortality, major
amputation, and decreased length of stay (Agarwal 2016).
Structured Exercise Current approaches for endovascular procedures include
balloon angioplasty, stents (covered and drug-eluting), and
Structured exercise is recommended for all symptomatic
atherectomy. In general, endovascular revascularization is
patients with PAD to improve quality of life and symptoms.
indicated when lifestyle-limiting claudication arises and sig-
General principles of structured exercise include walking until
nificant lesions are found on angiography (Gerhard-Herman
significant IC pain occurs, then briefly resting until the pain
2017).
subsides. Data consistently support the benefits of struc-
Despite high rates of restenosis, only minimal low-
tured exercise therapy in patients with symptomatic PAD. Pro-
level data exist regarding optimal antiplatelet therapy
grams can exist in hospital or outpatient facilities, as well as
post-intervention, which is a stark contrast from percutane-
within a structured community or home-based exercise pro-
ous coronary interventions from which much of the data are
gram. The sessions are typically, 30–45 minutes, three times
extrapolated. Dual antiplatelet therapy is recommended for
per week, for 12 weeks (Gerhard-Herman 2017).
at least 1 month by the 2017 ESC guidelines (class IIa, level
C) in patients who do not otherwise have an indication for
CRITICAL LIMB ISCHEMIA oral anticoagulation (Aboyans 2018). These guidelines were
In patients with PAD, CLI contributes substantially to mor- published before the results of the VOYAGER PAD trial, which
bidity and mortality. Adverse effects include pain, decreased demonstrated benefit with low-dose rivaroxaban after revas-
quality of life, immobility, and, ultimately, amputation and cularization (Bonaca 2020).
death. An analogous term, chronic limb-threatening ischemia,
encompasses the full spectrum of complications that arise Surgical Revascularization
from this condition, including impaired wound healing, neu- Surgical intervention should not be performed solely to pre-
ropathy, gangrene, and limb pain. All of these complications vent progression of disease. Primarily, surgery should be
contribute to the high morbidity and mortality rate associ- considered when a patient has progressed to CLI to pre-
ated with this advanced stage of disease. Long-term prog- vent amputation. In patients who have undergone surgi-
nosis once a diagnosis of CLI has been established is very cal bypass grafting, aspirin monotherapy improves graft
poor and associated with a fatality rate higher than many can- patency. For patients receiving a prosthetic graft, DAPT is
cers. Within 4 years of CLI diagnosis, mortality exceeds 50%; effective compared with aspirin alone (Aboyans 2018; Ger-
regardless of the revascularization approach, major amputa- hard-Herman 2017). With the recently published VOGAYER
tion rates range from 6%–10% (Mustapha 2019). PAD data, low-dose rivaroxaban plus aspirin should be con-
sidered for patients undergoing surgical or endovascular

PSAP 2022 Book 1 • Cardiology 72 Peripheral Arterial Disease


Asymptomatic Symptomatic Revascularization

Aspirin, or
Consider
clopidogrelb, Surgery Endovascular
aspirina
or ticagrelorc

Or

Low-dose Low-dose
DAPT for
rivaroxaband + rivaroxaband +
1–6 months
aspirine aspirine

Or/followed by

Low-dose
rivaroxaband +
aspirine,f

Figure 2. Approach to antithrombotic therapy in patients with PAD.


a
Recommended by the American Heart Association/American College of Cardiology guidelines.
b
Clopidogrel is preferred by European Society of Cardiology guidelines for single antiplatelet therapy.
c
Benefit with ticagrelor established in patients with history of percutaneous coronary stents.
d
Low-dose rivaroxaban is 2.5 mg twice daily.
e
Consider in patients with low bleeding risk.
f
Based on VOYAGER-PAD, limit overlap of 30-days of DAPT plus low-dose rivaroxaban.
DAPT = dual antiplatelet therapy; PAD = peripheral arterial disease.
Information from: Gerhard-Herman MD, Gornik HL, Barrett C, et al. 2016 AHA/ACC guideline on the management of patients with
lower extremity peripheral artery disease: a report of the American College of Cardiology/American Heart Association Task
Force on Clinical Practice Guidelines. J Am Coll Cardiol 2017;69:e71-126; Aboyans V, Ricco J, Bartelink M, et al. 2017 ESC
guidelines on the diagnosis and treatment of peripheral arterial diseases. Eur Heart J 2018;39:763-816; Bonaca MP, Bauersachs
RM, Anand SS, et al. Rivaroxaban in peripheral artery disease after revascularization. N Engl J Med 2020;382:1994-2004;
Hussain MA, Al-Omran M, Creager MA, et al. Antithrombotic therapy for peripheral artery disease: recent advances. J Am Coll
Cardiol 2018;71:2450-67.

revascularization (Bonaca 2020). Figure 2 provides a pro- atrial fibrillation or left ventricular mural thrombosis after MI.
posed treatment algorithm for patients with PAD. Shared decision-making should be incorporated when possi-
ble. Options for management include catheter-direct throm-
ACUTE LIMB ISCHEMIA bolysis and endovascular or surgical thrombectomy. Limbs
Acute limb ischemia is a medical emergency and should may not be salvageable upon presentation, and major ampu-
be recognized and treated rapidly. Commonly also referred tation may be necessary in cases of irreversible tissue loss.
to as cold leg, patients require rapid intervention to restore Urgent angiography, either by computed tomography or inva-
arterial blood flow. Lower extremity symptoms may include sively, is indicated when patients present with loss of sensa-
pain, loss of function or paresthesias, and coolness in the tion, paralysis, or lack of Doppler-detected blood flow (Fluck
affected extremity The acute nature is primarily caused by 2020; Gerhard-Herman 2017). Staging of ALI to make clinical
a rapid decrease in blood flow resulting from thrombosis or decisions can be done using the Rutherford classification
embolization. The source of embolization is primarily the (Table 3).
heart, aorta, or larger arteries and may be associated with
Anticoagulation

PSAP 2022 Book 1 • Cardiology 73 Peripheral Arterial Disease


Patient Care Scenario
A 72-year-old woman presents for follow-up after place- This patient should be receiving a high-intensity sta-
ment of a stent to the iliac artery 5 weeks ago. Today she tin (atorvastatin 40–80 mg/day or rosuvastatin 20–40
reports that her symptoms have improved. She is able to mg/day). Her LDL cholesterol should be rechecked in 12
walk her dog around the block and complete her grocery weeks, as recommended by the 2018 AHA/ACC guideline
shopping without stopping. Her history includes type 2 on management of blood cholesterol. To improve her A1C
DM, hypertension, and dyslipidemia. She recently quit control and further reduce her risk of CV events, adding
smoking. Relevant vital signs and laboratory values today a SGLT-2 inhibitor or a GLP-1 receptor agonist should be
are blood pressure 138/82 mm Hg, LDL cholesterol 115 considered. This patient is also not achieving her blood
mg/dL, and A1C 8.0%. Her current medications include pressure goal of less than 130/80 mm Hg. She should be
metformin 1000 mg twice daily, amlodipine 10 mg/day, receiving an ACEI or ARB to reduce her risk of future CV
pravastatin 80 mg/day at bedtime, aspirin 81 mg/day, and events. Based on the results of the VOYAGER PAD trial,
clopidogrel 75 mg/day. What recommendations should be this patient would benefit from the addition of rivaroxaban
made to optimize her medical regimen? 2.5 mg twice daily. Because it has been longer than 30
This patient presents with PAD with recent revascu- days from her surgical intervention, little data are avail-
larization. Her symptoms have improved; however, her able to support continuing clopidogrel as part of DAPT at
modifiable risk factors should continue to be addressed this time. However, the addition of low-dose rivaroxaban,
to reduce her risk of MACE and acute limb events. She in combination with aspirin, will reduce her risk of future
should be encouraged to maintain her smoking cessation. MACE and MALE.

1. Gerhard-Herman MD, Gornik HL, Barrett C, et al. 2016 AHA/ACC guideline on the management of patients with lower extremity peripheral
artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines.
J Am Coll Cardiol 2017;69:e71-126.
2. Aboyans V, Ricco J, Bartelink M, et al. 2017 ESC guidelines on the diagnosis and treatment of peripheral arterial diseases, in collabora-
tion with the European Society for Vascular Surgery. Eur Heart J 2018;39:763-816.
3. Bonaca MP, Bauersachs RM, Anand SS, et al. Rivaroxaban in peripheral artery disease after revascularization. N Engl J Med
2020;382:1994-2004.

Table 3. Rutherford Classification of Acute Limb Ischemia

Muscle Arterial Venous


Class Description Sensory Loss Weakness Doppler Doppler

I Viable Not immediately threatened None None Audible Audible

IIa Marginally Salvageable with prompt None or minimal None Absent Audible
threatened treatment (toes only)

IIb Immediately Salvageable with immediate More than toes; Mild to Absent Audible
threatened revascularization rest pain moderate

III Irreversible Major tissue loss, inevitable Profound Profound Absent Absent
nerve damage

Information from: Rutherford RB, Baker JD, Ernst C, et al. Recommended standards for reports dealing with lower extremity ischemia:
revised version. J Vasc Surg 1997;26:517–38.

Systemic anticoagulation should be initiated as soon as the rapid anticoagulant reversal. The goal of anticoagulation is
diagnosis of ALI is suspected. The most common agent for to prevent further thrombus propagation and to inhibit distal
management is intravenous unfractionated heparin using a thrombosis caused by decreased arterial flow. Although spe-
weight-based protocol (bolus 60–80 units/kg, followed by cific literature is lacking, it is reasonable to use alternative
12–18 units/kg/hour, titrated to goal activated partial throm- agents (bivalirudin or argatroban) in a patient with history of
boplastin time). Enoxaparin is not ideal because of the poten- heparin-induced thrombocytopenia (Patel 2013).
tial acute need for surgery or invasive procedures requiring

PSAP 2022 Book 1 • Cardiology 74 Peripheral Arterial Disease


Table 4. Catheter-Directed Thrombolysis Dosing for Box 3. Contraindications to Catheter-
Acute Limb Ischemia Directed Thrombolysis

Absolute contraindications
Thrombolytic Dose
• Absolute contraindication to anticoagulation
Alteplase 0.12–2 mg/hr; maximum, 40 mg total • Active clinically significant bleeding
Reteplase 0.25 to 1.0 U/h; maximum, 20 U in 24 hr
• Intracranial hemorrhage
• Presence/development of compartment syndrome
Tenecteplase Bolus infusion of 1–5 mg, followed by Relative contraindications
0.125–0.5 mg/hr • Bacterial endocarditis
• Bleeding diathesis
Information from: Patel NH, Krishnamurthy VN, Kim S, et al. • Cardiopulmonary resuscitation within past 10 days
Quality improvement guidelines for percutaneous manage- • Diabetic hemorrhagic retinopathy
ment of acute lower-extremity ischemia. J Vasc Interv • Disseminated intravascular coagulation
Radiol 2013;24:3-15; Ebben HP, Jongkind V, Wisselink W, et • Established cerebrovascular event (including transient
al. Catheter directed thrombolysis protocols for peripheral ischemic attacks) within past 2 mo
arterial occlusions: a systematic review. Eur J Vasc Endo- • Life expectancy < 1 yr
vasc Surg 2019;57:667-75. • Intracranial tumor, vascular malformation, aneurysm, or
seizure disorder
• Major surgery, or major trauma within past 10 days
• Neurosurgery (intracranial, spinal), or intracranial trauma
within past 3 mo
Catheter-Directed Strategies • Pregnancy and immediate postpartum status
Catheter-directed thrombolysis (CDT) in the management of • Recent eye surgery within past 3 mo
ALI has been associated with lower morbidity, decreased hos-
• Recent internal hemorrhage, puncture of noncompressible
vessel or organ biopsy
pital length of stay, and less patient discomfort. It is currently • Recent major GI bleeding within past 10 days
recommended when symptoms have been present for less • Serious allergic or other reaction to thrombolytic agent,
than 14 days. Current AHA/ACC guidelines state CDT is effec- anticoagulant, or contrast media (not controlled by steroid/
antihistamine pretreatment)
tive for patients with a salvageable limb (Gerhard-Herman
2017). There is no consensus regarding which fibrinolytic • Severe liver dysfunction, particularly in cases with
coagulopathy
agent or dosing regimen is optimal. Two general approaches
• Severe thrombocytopenia
are commonly used, although each has an associated • Uncontrolled hypertension (SBP >180 mm Hg or diastolic
risk: low-dose, long-duration thrombolytic infusions are blood pressure >110 mm Hg)
associated with risk of hemorrhage and high-dose, and
Information from: Patel NH, Krishnamurthy VN, Kim S, et al.
short-duration infusions may increase risk of distal embo- Quality improvement guidelines for percutaneous manage-
lization. Most institutions design dosing regimens with ment of acute lower-extremity ischemia. J Vasc Interv Radiol
short-durations (between 18–22 hours) and attempt to min- 2013;24:3-15.

imize overall drug exposure. Suggested dosing is outlined in


Table 4 (Patel 2013).
A recent systematic review included 9877 patients who
received CDT in the lower extremities. The mean treatment
duration was 21.4 hours (95% CI, 21.0–21.8), with 82% of pro- future amputation (Fluck 2020). Hybrid CDT can also be used
tocols reporting bolus dosing. Overall angiographic patency in combination with aspiration or mechanical thrombectomy
was achieved in 75% (95% CI, 74.6–75.1) of patients. No ampu- to facilitate restoration of blood flow (see Table 4). Pharmaco-
tation was performed for 91% (95% CI, 90.3 - 90.7) of patients. therapy after CDT is dependent on the underlying lesion. If no
Bleeding complications occurred in 18% (95% CI, 17.8–18.3), endovascular or surgical intervention is necessary, patients
which remains the primary concern regarding this approach should receive anticoagulation (Patel 2013).
(Ebben 2019). The Society of Interventional Radiology main-
tains quality improvement guidelines that include absolute
and relative contraindications to thrombolytics. Authors note CONCLUSION
this list was reached by consensus, rather than literature eval- Peripheral arterial disease continues to be underdiagnosed
uation. The suggested contraindications (Box 3) should be and undertreated despite the high morbidity and mortality
considered and discussed with patients as part of a shared associated with this disease. Major gaps in data related to
decision-making process (Patel 2013). appropriate treatment exist. However, advances have been
Patients who undergo CDT benefit from subsequent sur- made with the COMPASS and VOYAGER PAD trials. Patients
gical or endovascular revascularization to decrease need for presenting with ALI require emergency management to

PSAP 2022 Book 1 • Cardiology 75 Peripheral Arterial Disease


artery disease: an international, randomised, double-blind,
Practice Points placebo-controlled trial. Lancet 2018;391:219-29.
• Clinically, PAD is considered ASCVD, and mitigation of CV
risk factors is important to reduction of disease progres- Antithrombotic Trialists’ Collaboration. Collaborative
sion and ischemic events. meta-analysis of randomised trials of antiplatelet therapy
• Early detection by appropriate screening and aggressive for prevention of death, myocardial infarction, and stroke
risk reduction strategies are crucial to prevent disease in high risk patients. BMJ. 2002 Jan 12;324(7329):71-86.
progression and mortality.
Armstrong EJ, Wu J, Singh GD, et al. Smoking ces-
• Treatment is primarily focused on patients with symptom-
sation is associated with decreased mortality and
atic PAD with limited data on screening patients who are
improved amputation-free survival among patients with
truly asymptomatic.
symptomatic peripheral artery disease. J Vasc Surg
• Smoking cessation should be offered at every office visit
2014;60:1565-71.
for current smokers.
• Aspirin, clopidogrel, statins, evolocumab, rivaroxaban and Bavry AA, Anderson RD, Gong Y, et al. Outcomes among
ACEI/ARBs all reduce the risk of MACE in patients with hypertensive patients with concomitant peripheral
symptomatic PAD. and coronary artery disease: findings from the Interna-
• Rivaroxaban reduced the risk of MACE and MALE after tional Verapamil-SR/Trandolapril Study. Hypertension
revascularization, regardless of short-term clopidogrel use. 2010;55:48-53.
• Patients with ALI have a very high morbidity and mortality,
and this condition should be managed as an emergency, Barua RS, Rigotti NA, Benowitz NL, et al. 2018 ACC expert
including intravenous anticoagulation. consensus decision pathway on tobacco cessation treat-
• The use of CDT can offer an alternative to surgery in ment: a report of the American College of Cardiology Task
patients with ALI; however, this approach carries a risk of Force on Clinical Expert Consensus Documents. J Am Coll
bleeding, and dosing strategies lack standardization. Cardiol 2018;72:3332-65.

Bedenis R, Stewart M, Cleanthis M, et al. Cilostazol for


intermittent claudication. Cochrane Database Syst Rev
increase likelihood of limb salvage. Pharmacists are well 2014:CD003748.
positioned to advocate for patients with PAD, to address mod- Bonaca MP, Bauersachs RM, Anand SS, et al. Rivaroxaban in
ifiable risk factors, and to initiate pharmacotherapy to lower peripheral artery disease after revascularization. N Engl J
CV risk. Med 2020;382:1994–2004.

Bonaca MP, Bhatt DL, Storey RF, et al. Ticagrelor for pre-
REFERENCES vention of ischemic events after myocardial infarction in
Aboyans V, Criqui MH, Abraham P, et al. Measurement and patients with peripheral artery disease. J Am Coll Cardiol
interpretation of the ankle-brachial index: a scientific 2016;67:2719-28.
statement from the American Heart Association. Circula-
tion 2012;126:2890-909. Bonaca MP, Nault P, Giugliano RP, et al. Low-density lipo-
protein cholesterol lowering with evolocumab and out-
Aboyans V, Ricco J, Bartelink M, et al. 2017 ESC guidelines comes in patients with peripheral artery disease: insights
on the diagnosis and treatment of peripheral arterial dis- from the FOURIER trial (further cardiovascular outcomes
eases, in collaboration with the European Society for Vas- research with PCSK9 inhibition in subjects with elevated
cular Surgery (ESVS). Eur Heart J 2018;39:763-816. risk). Circulation 2018;137:338-50.

Agarwal S, Sud K, Shishehbor MH. Nationwide trends of Bonaca MP, Scirica BM, Creager MA, et al. Vorapaxar in
hospital admission and outcomes among critical limb patients with peripheral artery disease: results from
ischemia patients: from 2003–2011. J Am Coll Cardiol TRA2{degrees}P-TIMI 50. Circulation 2013;127:1522-9.
2016;67:1901-13.
Brown T, Forster RB, Cleanthis M, et al. Cilostazol for inter-
American Diabetes Association. 9. Pharmacologic mittent claudication. Cochrane Database Syst Rev
approaches to glycemic treatment: standards of medical 2021:CD003748.
care in diabetes—2021. Diabetes Care 2021a;44:S111-24.
Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added
American Diabetes Association. 10. Cardiovascular disease to statin therapy after acute coronary syndromes. N Engl J
and risk management: standards of medical care in diabe- Med 2015;372:2387-97.
tes—2021. Diabetes Care 2021b;44:S125-50.
CAPRIE Steering Committee. A randomised, blinded, trial of
Anand S, Yusuf S, Xie C, et al; Warfarin Antiplatelet Vascular clopidogrel versus aspirin in patients at risk of ischaemic
Evaluation (WAVE) Trial Investigators. Oral anticoagulant events (CAPRIE). CAPRIE Steering Committee. Lancet
and antiplatelet therapy and peripheral arterial disease. N 1996;348:1329-39.
Engl J Med 2007;357:217-27.
Cacoub PP, Bhatt DL, Steg PG, at al. CHARISMA Investiga-
Anand SS, Bosch J, Eikelboom JW, et al. Rivaroxaban with or tors. Patients with peripheral arterial disease in the CHA-
without aspirin in patients with stable peripheral or carotid RISMA trial. Eur Heart J. 2009;2:192-201.

PSAP 2022 Book 1 • Cardiology 76 Peripheral Arterial Disease


Dawson DL, Cutler BS, Hiatt WR, et al. A comparison of Hirsch AT, Criqui MH, Treat-Jacobson D, et al. Peripheral
cilostazol and pentoxifylline for treating intermittent clau- arterial disease detection, awareness, and treatment in
dication. Am J Med 2000;109:523-30. primary care. JAMA 2001;286:1317-24.

Ding N, Sang Y, Chen J, et al. Cigarette smoking, smoking Jukema JW, Szarek M, Zijlstra LE, et al. Alirocumab in
cessation, and long-term risk of 3 major atherosclerotic patients with polyvascular disease and recent acute coro-
diseases. J Am Coll Cardiol 2019;74:498-507. nary syndrome: ODYSSEY OUTCOMES trial. J Am Coll Car-
diol 2019;74:1167-76.
Ebben HP, Jongkind V, Wisselink W, et al. Catheter directed
thrombolysis protocols for peripheral arterial occlu- Kumbhani DJ, Steg PG, Cannon CP, et al. Statin therapy and
sions: a systematic review. Eur J Vasc Endovasc Surg long-term adverse limb outcomes in patients with periph-
2019;57:667-75. eral artery disease: insights from the REACH registry. Eur
Heart J 2014;35:2864-72.
Eikelboom JW, Connolly SJ, Bosch J, et al. Rivaroxaban with
or without aspirin in stable cardiovascular disease. N Engl Lee HF, Chen SW, Liu JR, et al. Major adverse cardiovascu-
J Med 2017;377:1319-30. lar and limb events in patients with diabetes and concomi-
tant peripheral artery disease treated with sodium glucose
Espinola-Klein C, Weisser G, Jagodzinski A, et al. β-Blockers cotransporter 2 inhibitor versus dipeptidyl peptidase-4
in patients with intermittent claudication and arterial inhibitor. Cardiovasc Diabetol 2020;19:160.
hypertension: results from the nebivolol or metopr-
olol in arterial occlusive disease trial. Hypertension Leng GC, Fowkes FG. The Edinburgh Claudication Question-
2011;58:148-54. naire: an improved version of the WHO/Rose question-
naire for use in epidemiological surveys. J Clin Epidemiol
Fluck F, Augustin AM, Bley T, et al. Current treatment options 1992;45:1101-09.
in acute limb ischemia. Rofo 2020;192:319-26.
Maffei A, Lembo G. Nitric oxide mechanisms of nebivolol.
Fowkes FG, Price JF, Stewart MC, et al. Aspirin for prevention Ther Adv Cardiovasc Dis 2009; 4:317-27.
of cardiovascular events in a general population screened
for a low ankle brachial index: a randomized controlled McDermott MM, Kerwin DR, Liu K, et al. Prevalence and sig-
trial. JAMA 2010;303:841-8. nificance of unrecognized lower extremity peripheral arte-
rial disease in general medicine practice. J Gen Intern
Gerhard-Herman MD, Gornik HL, Barrett C, et al. 2016 AHA/ Med. 2001; 6:384-90.
ACC guideline on the management of patients with lower
extremity peripheral artery disease: a report of the Amer- Moll F, Baumgartner I, Jaff M, et al. Edoxaban plus aspirin
ican College of Cardiology/American Heart Association vs dual antiplatelet therapy in endovascular treatment
Task Force on Clinical Practice Guidelines. J Am Coll Car- of patients with peripheral artery disease: results of the
diol 2017;21;69:e71-126. ePAD trial. J Endovasc Ther 2018;25:158-68.

Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/ Montgomery PS, Gardner AW. The clinical utility of a six-min-
AACVPR/AAPA/ABC/ACPM/ ADA/AGS/ APhA/ASPC/NLA/ ute walk test in peripheral arterial occlusive disease
PCNA guideline on the management of blood cholesterol: patients. J Am Geriatr Soc 1998;46:706-11.
a report of the American College of Cardiology/American
Heart Association Task Force on Clinical Practice Guide- Morrow DA, Braunwald E, Bonaca MP, et al. Vorapaxar in the
lines. J Am Coll Cardiol 2019;73:e285-350. secondary prevention of atherothrombotic events. N Engl
J Med 2012;366:1404-13.
Henry AJ, Hevelone ND, Belkin M, Nguyen LL. Socioeco-
nomic and hospital-related predictors of amputation for Mustapha JA, Katzen BT, Neville RF, et al. Propensity
critical limb ischemia. J Vasc Surg 2011;53:330-9.e.1. score-adjusted comparison of long-term outcomes among
revascularization strategies for critical limb ischemia. Circ
Hiatt WR, Bonaca MP, Patel MR, et al. Rivaroxaban and aspi- Cardiovasc Interv 2019;12:e008097.
rin in peripheral artery disease lower extremity revascular-
ization: impact of concomitant clopidogrel on efficacy and Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and car-
safety. Circulation 2020;142:2219-30. diovascular and renal events in type 2 diabetes. N Engl J
Med 2017;377:644-57.
Hiatt WR, Fowkes FG, Heizer G, et al. Ticagrelor versus clopi-
dogrel in symptomatic peripheral artery disease. N Engl J ONTARGET Investigators, Yusuf S, Teo KK, et al. Telmisar-
Med 2017;376:32-40. tan, ramipril, or both in patients at high risk for vascular
events. N Engl J Med 2008;358:1547-59.
Hira RS, Cowart JB, Akeroyd JM, et al. Risk factor optimiza-
tion and guideline-directed medical therapy in US Veter- Ostergren J, Sleight P, Dagenais G, et al. Impact of ramipril in
ans with peripheral arterial and ischemic cerebrovascular patients with evidence of clinical or subclinical peripheral
disease compared to Veterans with coronary heart dis- arterial disease. Eur Heart J 2004;25:17-24.
ease. Am J Cardiol 2016;118:1144-9. Patel NH, Krishnamurthy VN, Kim S, et al. Quality improve-
ment guidelines for percutaneous management of
acute lower-extremity ischemia. J Vasc Interv Radiol
2013;24:3-15.

PSAP 2022 Book 1 • Cardiology 77 Peripheral Arterial Disease


Shu J, Santulli G. Update on peripheral artery disease: epi-
demiology and evidence-based facts. Atherosclerosis
2018;275:379-81.

Stein R, Hriljac I, Halperin JL, et al. Limitation of the resting


ankle-brachial index in symptomatic patients with periph-
eral arterial disease. Vasc Med 2006;11:29-33.

Verma S, Mazer CD, Al-Omran M, et al. Cardiovascular out-


comes and safety of empagliflozin in patients with type 2
diabetes mellitus and peripheral artery disease: a subanal-
ysis of EMPA-REG OUTCOME. Circulation 2018;137:405-7.

Virani SS, Alonso A, Benjamin EJ, et al. Heart disease and


stroke statistics—2020 update: a report from the American
Heart Association. Circulation 2020;141:e139-596.

Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/


AAPA/ABC/ACPM/AGS/APhA/ASH/ ASPC/NMA/PCNA
guideline for the prevention, detection, evaluation, and
management of high blood pressure in adults: a report of
the American College of Cardiology/American Heart Asso-
ciation Task Force on Clinical Practice Guidelines. Hyper-
tension 2018;71:e13-115.

PSAP 2022 Book 1 • Cardiology 78 Peripheral Arterial Disease


Self-Assessment Questions
1. A 50-year-old African American man (weight 98 kg Questions 4–6 pertain to the following case.

[216 lb], height 70 inches) has a medical history that A.Y., a 65-year-old woman with a medical history of T2DM
includes type 2 diabetes mellitus (T2DM), hypertension, (A1C 8.1%), stable ischemic heart disease (SIHD), and hyper-
and hyperlipidemia. He quit smoking 25 years ago and tension presents to the clinic with a chief complaint of wors-
currently drinks 1–2 beers per day. The patient reports ening leg cramping and fatigue that increases on her daily
pain and cramping in his calves when walking his dog walks and is relieved by rest. She notes that compared with
around the block. The pain subsides when he rests on a 6 months ago, she can no longer walk as far as she used to,
park bench. Physical examination reveals loss of leg hair especially in the past month. A.Y. has smoked 1 pack of ciga-
and shiny calves with diminished pulses. Ankle-brachial rettes per day for the past 40 years. Upon physical examina-
index (ABI) are measured at 0.65 on the right leg and 0.72 tion her legs are cool to the touch but pulses are palpable. Her
on the left leg. Which one of the following is this patient’s home drugs include metformin 1000 mg twice daily, lisinopril
greatest risk factor for major amputation? 20 mg/day, and aspirin 81 mg/day.

A. Obesity 4. Based on her symptoms and physical examination, A.Y.


B. Race is referred for ABI evaluation. The patient’s systolic blood
C. Hypertension pressure (SBP) measurements are recorded as the fol-
D. Age lowing: 136 mm Hg (brachial, right arm), 142 mm Hg (bra-
chial, left arm), 110 mm Hg (dorsalis pedis, right foot), 118
2. A 61-year-old woman with a medical history of T2DM
mm Hg (posterior tibial, right foot), 120 mm Hg (dorsa-
(A1C 7.2%), hypertension, and hyperlipidemia presents
lis pedis, left foot), and 122 mm Hg (posterior tibial, left
with complaints of lower extremity pain when walking
foot). Given these measurements, which one of the fol-
short distances. The pain resolves with rest. The patient
lowing best evaluates A.Y.’s left and right ABIs?
is a current 1 pack per day smoker. Testing reveals ABIs
of 0.80 in both legs with no visible skin ulcers or gan- A. 0.88 (left) and 0.77 (right)
grene. Today’s vital signs include blood pressure 122/74 B. 0.84 (left) and 0.86 (right)
mm Hg and heart rate 78 beats/minute. Her home drugs C. 0.86 (left) and 0.83 (right)
include amlodipine 10 mg/day, rosuvastatin 20 mg/day, D. 0.81 (left) and 0.81 (right)
lisinopril 20 mg/day, metformin 1000 mg twice daily, and 5. To optimize her diabetes regimen by providing cardiovas-
semaglutide 1 mg subcutaneously weekly. Which one cular (CV) risk reduction, which one of the following is
of the following is best to recommend for this patient best to recommend for A.Y.?
to decrease her risk of a major adverse cardiovascular
A. Insulin
event (MACE)?
B. Empagliflozin
A. Increase lisinopril to 40 mg/day. C. Glipizide
B. Initiate aspirin 81 mg/day. D. Exenatide
C. Initiate cilostazol 100 mg twice daily.
6. Which one of the following is best to recommend to
D. Initiate warfarin (INR goal 2–3).
reduce A.Y.’s risk of CV death and acute limb events?
3. A 71-year-old woman with established PAD (previous
A. Clopidogrel
ABIs 0.87 and 0.88) was initiated on atorvastatin
B. Cilostazol
80 mg/day 4 months ago. Today she returns to the office
C. Rivaroxaban
for follow-up. The patient has no history of previous
D. Amlodipine
peripheral revascularization. Her LDL cholesterol today
is 82 mg/dL. Which one of the following is best to recom-
Questions 7 and 8 pertain to the following case.
mend to optimize this patient’s medical regimen?
K.T., a 69-year-old man, is preparing for discharge from
A. Maintain current therapy.
the hospital after placement of a drug-eluting stent (DES)
B. Add ezetimibe.
to his femoral artery due to chronic limb ischemia with a
C. Change to rosuvastatin 40 mg/day.
non-healing ulcer on his left foot. His medical history includes
D. Add evolocumab.
coronary artery disease, hypertension, and hyperlipidemia.

PSAP 2022 Book 1 • Cardiology 79 Peripheral Arterial Disease


K.T. has smoked one pack per day for the past 45 years. His and ejection fraction (EF) is 52% (on ECHO). Which one of
drugs prior to the procedure included aspirin 81 mg/day, tel- the following is best to recommend to optimize manage-
misartan 80 mg/day, rosuvastatin 20 mg/day, and amlodipine ment of this patient’s PAD?
10 mg/day.
A. Initiate evolocumab 420 mg subcutaneously
7. Which one of the following is best to recommend to monthly.
decrease K.T.’s risk of acute limb events? B. Discontinue cilostazol 100 mg twice daily.
C. Increase lisinopril to 20 mg/day.
A. Low-dose rivaroxaban plus low-dose aspirin
D. Change aspirin to rivaroxaban 2.5 mg twice daily.
B. Dual antiplatelet therapy (DAPT) for 30 days, then
aspirin 325 mg/day indefinitely 11. You are developing an institutional protocol for catheter-
C. Low-dose aspirin monotherapy directed thrombolysis for patients presenting with acute
D. Warfarin (INR goal 2–3) limb ischemia. Alteplase is the thrombolytic on formu-
lary. Which one of the following dosing protocols best
8. A 59-year-old white man with a medical history that
balances the risks and benefits in this patient population?
includes ST-segment elevation myocardial infarction
12 months ago, PAD, hypertension, and hyperlipidemia A. 2 mg/hour for 48 hours
is seen today for his 1-year follow-up. His home drugs B. 1 mg/hour for 5 hours
include aspirin 81 mg/day, lisinopril 40 mg/day, ator- C. 1 mg/hour for 18 hours
vastatin 80 mg/day, metoprolol succinate 100 mg/day, D. 0.5 mg/hour for 10 hours
amlodipine 10 mg/day, and ticagrelor 90 mg twice daily.
12. A 68-year-old man with medical history of T2DM, hyper-
Upon physical examination his blood pressure is 102/64
tension, coronary artery disease, and heart failure with
mm Hg and heart rate is 71 beats/minute. His home
reduced EF presents to the ED with worsening leg pain.
blood pressure logs are consistent with the office mea-
Upon examination, his foot is cool to the touch and a
surement. Which one of the following is best to recom-
large non-healing ulcer is noted. Dopplers are absent.
mend to manage this patient’s blood pressure?
The patient undergoes femoropopliteal bypass surgery.
A. Increase metoprolol succinate to 150 mg/day. Which one of the following antithrombotic regimens is
B. Discontinue amlodipine. best to recommend to reduce this patient’s risk of recur-
C. Discontinue lisinopril. rence of CV death and future limb events?
D. Continue current therapy.
A. Low-dose rivaroxaban plus low-dose aspirin
9. A 68-year-old man with previously documented PAD, B. DAPT for 30 days, then aspirin 81 mg/day
T2DM, and hypertension is seen in clinic for follow-up. indefinitely
He has participated in a structured exercised program C. Low-dose aspirin monotherapy
with some improvement in his symptoms; however, D. Warfarin (INR goal 2–3)
he still feels his quality of life is limited by symptoms.
His current drugs include aspirin 81 mg/day, atorvas- Questions 13–15 pertain to the following case.
tatin 40 mg/day, losartan 100 mg/day, and metformin R.A., a 73-year-old man (weight 98 kg [216 lb], CrCl 58 mL/min)
500 mg twice daily. Which one of the following is best with a medical history of T2DM, hypertension, and hyperlipid-
to recommend to address this patient’s symptoms of emia presents to the ED with 2 days of worsening leg pain.
claudication? This morning his leg became increasingly painful at rest and
A. Cilostazol he has lost sensation in his toes. R.A.’s arterial Dopplers are
B. Pentoxifylline absent, but venous Dopplers are audible. He reports no mus-
C. Clopidogrel cle weakness at this time.
D. Ticagrelor 13. Given his current symptoms, which one of the follow-
10. A 66-year-old woman with a medical history of PAD pres- ing anticoagulation strategies is best to recommend for
ents to her cardiologist for follow-up. Her current home R.A.?
drugs include: aspirin 81 mg/day, cilostazol 100 mg A. Intravenous heparin
twice daily, lisinopril 10 mg/day, atorvastatin 80 mg/day, B. Enoxaparin 100 mg subcutaneously every 12 hours
and ezetimibe 10 mg/day. The patient reports improve- C. Warfarin (INR 2–3)
ment in her symptoms since cilostazol was started 6 D. Rivaroxaban 2.5 mg twice daily
months ago. Pertinent objective data include blood pres-
sure 128/74 mm Hg, heart rate 88 beats/minute, and LDL
cholesterol 90 mg/dL; ECG shows normal sinus rhythm,

PSAP 2022 Book 1 • Cardiology 80 Peripheral Arterial Disease


14. Based on R.A.’s presentation and additional imaging, he 15. After catheter-directed thrombolysis, R.A. undergoes
is scheduled to undergo catheter-directed thrombolysis. peripheral angiography and receives a DES to superficial
Balancing dosing regimens and principles, which one of femoral artery. In addition to aspirin 81 mg/day, which
the following thrombolytic regimens is best to recom- one of the following antithrombotic regimens is best to
mend for R.A.? recommend for 30 days after R.A.’s procedure?
A. Tenecteplase 2 mg bolus, followed 0.25 mg/hour A. Warfarin (INR 2-3)
infusion for 18 hours B. Ticagrelor 60 mg twice daily
B. Alteplase 2 mg/hour infusion for 36 hours C. Clopidogrel 75 mg/day
C. Tenecteplase 0.125 mg/hour infusion for 36 hours D. Apixaban 2.5 mg twice daily
D. Reteplase 1 U/hour infusion for 24 hours

PSAP 2022 Book 1 • Cardiology 81 Peripheral Arterial Disease


Learner Chapter Evaluation: Peripheral Arterial Disease

As you take the posttest for this chapter, also evaluate the 8. The teaching and learning methods used in the chapter
material’s quality and usefulness, as well as the achievement were effective.
of learning objectives. Rate each item using this 5-point scale: 9. The active learning methods used in the chapter were
effective.
• Strongly agree
10. The learning assessment activities used in the chapter
• Agree
were effective.
• Neutral
• Disagree 11. The chapter was effective overall.
• Strongly disagree 12. The activity met the stated learning objectives.
13. If any objectives were not met, please list them here.
1. The content of the chapter met my educational needs.
2. The content of the chapter satisfied my expectations.
3. The author presented the chapter content effectively.
OTHER COMMENTS
4. The content of the chapter was relevant to my practice 14. Please provide any specific comments related to any
and presented at the appropriate depth and scope. perceptions of bias, promotion, or advertisement of
commercial products.
5. The content of the chapter was objective and balanced.
15. Please expand on any of your above responses, and/or
6. The content of the chapter is free of bias, promotion, and
provide any additional comments regarding this chapter:
advertisement of commercial products.
7. The content of the chapter was useful to me.

PSAP 2022 Book 1 • Cardiology 82 Peripheral Arterial Disease


Antithrombotic Therapy in Cardiac
Interventions
By Stephanie Dwyer Kaluzna, Pharm.D., BCCP; and Jaclynne Gowen, Pharm.D., BCCP

Reviewed by A. Josh Roberts, Pharm.D., BCCP, BCPS-AQ Cardiology, AACC: Bethany A. Ford, Pharm.D., BCPS; and Christine S. Ji, Pharm.D.,
BCCP, BCPS

LEARNING OBJECTIVES

1. Apply the advances made in percutaneous coronary intervention and complications with coronary stenting to determine
appropriate antithrombotic regimens.
2. Analyze recent literature surrounding the use of chemoreceptor P2Y12 inhibitors and oral anticoagulants in patients with
coronary artery disease and acute coronary syndromes.
3. Design a treatment plan for a patient with an indication for chronic anticoagulation who is undergoing percutaneous
coronary intervention.
4. Distinguish between the various antithrombotic treatment regimens indicated for patients undergoing transcatheter
valvular interventions.

PERCUTANEOUS CORONARY
ABBREVIATIONS IN THIS CHAPTER
INTERVENTION
ACS Acute coronary syndrome
Overview of Procedure and Indications
AS Aortic stenosis
CABG Coronary artery bypass graft Coronary artery disease (CAD) is a pathological process charac-
CAD Coronary artery disease terized by atherosclerotic plaque accumulation in the epicardial
DAPT Dual antiplatelet therapy arteries. It is categorized as either obstructive or nonobstructive
DAT Dual antithrombotic therapy depending on the degree of blockade within the coronary arteries.
DES Drug-eluting stent The disease state can have lengthy, stable periods, but it can become
DOAC Direct oral anticoagulant unstable at any time. An unstable presentation is most often caused
ESC European Society of Cardiology by an acute, atherothrombotic event caused by plaque rupture or ero-
sion. Depending on the nature of the CAD process, various clinical
ISR In-stent restenosis
presentations may emerge as either an acute coronary syndrome
NSTE-ACS Non-ST-elevation acute coronary
syndrome (ACS) or stable CAD, also known as stable ischemic heart disease
OAC Oral anticoagulation (SIHD). Invasive interventions such as percutaneous coronary inter-
PCI Percutaneous coronary vention (PCI) may be conducted to treat acute symptoms and prevent
intervention disease progression (Knuuti 2020). When a patient undergoes PCI, a
SAPT Single antiplatelet therapy coronary stent is typically placed at the lesion site to restore blood
SIHD Stable ischemic heart disease flow and prevent further myocardial ischemia.
ST Stent thrombosis After PCI, dual antiplatelet therapy (DAPT) is standard to prevent
STEMI ST-segment-elevation myocardial coronary thrombotic complications (Levine 2016). The term DAPT
infarction refers to a combination of aspirin—usually less than 100 mg daily—
TAT Triple antithrombotic therapy and a P2Y12 inhibitor. In the United States, three common oral P2Y12
TAVI Transcatheter aortic valvular inhibitors are FDA approved for use after PCI: clopidogrel, ticagre-
implantation lor, and prasugrel. Recently, intravenous P2Y12 inhibitor cangrelor
VKA Vitamin K antagonist became approved for use during PCI. Ticagrelor and prasugrel are
often referred to as novel P2Y12 inhibitors because they came to mar-
Table of other common abbreviations.
ket after clopidogrel and DAPT had become established post-PCI.
Both prasugrel and ticagrelor offer reductions in thrombotic events

PSAP 2022 Book 1 • Cardiology 83 Antithrombotic Therapy in Cardiac Interventions


based on increased platelet inhibition, but the reductions that influence P2Y12 inhibitor selection include indication for
come with an increased risk of bleeding (Levine 2016; Wal- PCI (i.e., SIHD versus ACS), adverse-effect profiles, cost,
lentin 2009; Wiviott 2007). It is important to note that factors drug interactions, and patient-specific ischemic and bleed-
ing risk factors. Table 1 summarizes the pertinent pharma-
cokinetic and pharmacodynamic properties of the P2Y12
inhibitors.
BASELINE KNOWLEDGE STATEMENTS
Evolution of Stent Types
Readers of this chapter are presumed to be familiar
Coronary stents have evolved significantly since their intro-
with the following:
duction in the mid-1980s, and new features that refine their
• General knowledge of the pathophysiology that design, structure, and material continue to emerge. Patient
leads to acute coronary syndromes and valvular
heart disease factors such as bleeding risk, thrombotic risk, lesion charac-
teristics, and presenting symptoms (SIHD vs. ACS) are con-
• General knowledge of coronary angiography, per-
cutaneous coronary intervention, transcatheter sidered when the type of stent is getting selected because
aortic valve implantation, and MitraClip insertion recommendations for antiplatelet therapies and length of
• General pharmacologic knowledge of P2Y 12
inhibi- therapy vary depending on stent type (Iqbal 2013).
tors and oral anticoagulants used in the treatment Bare-metal stents (BMSs) have historically used stainless
of coronary disease, atrial fibrillation, and valvular steel or cobalt-chromium. Most recently, platinum-chromium
heart disease alloy metals produce thinner struts without compromising
strength or conformability. An incidence of in-stent rest-
Table of common laboratory reference values
enosis (ISR) of 20%–30% has been reported with a BMS,
which is associated with significant morbidity and mortal-
ADDITIONAL READINGS
ity. Drug-eluting stents (DESs) were developed to specifically
The following free resources have additional back- correct ISR complications encountered with BMSs; they con-
ground information on this topic: tain antiproliferative drugs that reduce the development of
ISR. Second-generation DESs contain everolimus or zotar-
• Levine GN, Bates ER, Bittl JA, et al. 2016 ACC/AHA
guideline focused update on duration of dual anti- olimus and proved superiority in efficacy and safety com-
platelet therapy in patients with coronary artery pared with first-generation stents containing paclitaxel or
disease. J American Coll Cardiol 2016;68:1082-115. sirolimus (Iqbal 2013). The antiproliferative drugs are incor-
• Angiolillo DJ, Bhatt DL, Cannon CP, et al. Antithrom- porated within a polymer and coated on the surface of the
botic therapy in patients with atrial fibrillation stent; then they are released slowly over several weeks after
treated with oral anticoagulation undergoing percu- stent deployment.
taneous coronary intervention: a north american
The evolution of DESs reduced the rate of ISR, but
perspective: 2021 update. Circulation.
2021;143:583-96. increased rates of stent thrombosis (ST) resulted. In par-
ticular, late ST secondary to compromised polymer durabil-
• ten Berg J, Sibbing D, Rocca B, et al. Management ity remains of concern, and therefore, novel bioresorbable
of antithrombotic therapy in patients undergoing
transcatheter aortic valve implantation: a consen- stents were developed that are made up of metallic alloy or
sus document of the ESC working group on throm- polymer—with or without antiproliferative drugs. The ratio-
bosis and the european association of nale for a bioresorbable stent is to provide a scaffold, similar
percutaneous cardiovascular interventions to a stent, for a specific period of time after PCI, with grad-
(EAPCI), in collaboration with the ESC council on
ual resorption over time. After resorption, the vessel wall may
valvular heart disease. European Heart Journal.
regain normal function without the presence of foreign mate-
2021;42:2265-9.
rials, thus reducing the risk of ST and the requirement for
• Cahill TJ, Chen M, Hayashida K, et al. Transcatheter long-term DAPT (Iqbal 2013).
aortic valve implantation: current status and future
perspectives. European Heart Journal
2018;39:2625-34. Complications with Stents

• Hensey M, Brown RA, Lal S, et al. Transcatheter Two complications associated with coronary stent placement
mitral valve replacement. JACC: Cardiovascular are ST and ISR. Stent thrombosis is usually an acute process
Interventions. 2021;14:489-500. involving a thrombotic occlusion of a coronary stent. The
• Patrono C, Morais J, Baigent C, et al. Antiplatelet complication has been significantly reduced by implemen-
agents for the treatment and prevention of coro- tation of DAPT with aspirin plus a P2Y12 inhibitor after stent
nary atherothrombosis. Journal of the American placement (Neumann 1996). In contrast, ISR is a gradual nar-
College of Cardiology 2017;70:1760-76.
rowing of the stent lumen that is caused by neointimal prolif-
eration and has been improved with the development of DES.

PSAP 2022 Book 1 • Cardiology 84 Antithrombotic Therapy in Cardiac Interventions


Table 1. Pharmacokinetic and Pharmacodynamic Factors of P2Y12 Inhibitors

PK/PD Cangrelor (IV) Clopidogrel (PO) Prasugrel (PO) Ticagrelor (PO)

Dosing Load: 30 mcg/kg Load: 300–600 mg Load: 60 mg Load: 180 mg


Maintenance: Maintenance: 75 mg Maintenance: 10 mg Maintenance: 90 mg
4 mcg/kg/min daily daily (5 mg if weight twice daily
less than 60 kg)a

Pertinent pharmaco- Dephosphorylation to Prodrug Prodrug Hepatic via CYP3A4/5


kinetics inactive metabolite Esterase-mediated Rapid hydrolysis, to active metabolite
hydrolysis and CYP450-mediated
CYP450-mediated oxidation (CYP3A4
oxidation (CYP2C19) and CYP2B6)

Receptor blockade Reversible Irreversible Irreversible Reversible

Onset of action 2 minutes 2–6 hours ~30 minutes 30 minutes to 2 hours


Peak within 4 hours

Duration of Platelet function 5–10 days 7–10 days 3–5 days


antiplatelet effects normalizes within 1 hour
of discontinuation

a
Even though the FDA approved prasugrel for 5 mg daily in patients with body weights of less than 60 kg, the approval is based primar-
ily on pharmacokinetic data (Erlinge 2012). There are limited clinical efficacy and safety data with this dosing recommendation.
Information from: Brilakis ES, Patel VG, Banerjee S. Medical management after coronary stent implantation: a review. JAMA
2013;310:189-98; Baron TH, Kamath PS, McBane RD. Current concepts: management of antithrombotic therapy in patients
undergoing invasive procedures. N Engl J Med 2013;368:2113-24; Gurbel PA, Bliden KP, Butler K, et al. Randomized double-blind
assessment of the ONSET and OFFSET of the antiplatelet effects of ticagrelor versus clopidogrel in patients with stable coronary
artery disease: the ONSET/OFFSET study. Circulation 2009;120:2577-85.

Stent Thrombosis Thankfully, the introduction of DESs has drastically reduced


Stent thrombosis could occur at any time after coronary stent ISR (Stefanini 2013), which occurred up to three times more
placement and is strongly associated with mortality and myo- often with BMS compared with new-generation DESs within
cardial infarction (Guerra 2014). The timing of ST is catego- 12 months (Taniwaki 2014).
rized as early (within 30 days), which is further divided into
acute (within 24 hours) and subacute (days 2–30); late (from
USE OF P2Y12 INHIBITORS IN
31 days to 1 year); and very late (beyond 1 year) (Valgimigli
PERCUTANEOUS CORONARY
2018). Factors leading to early ST include stent underexpan-
INTERVENTION
sion, incomplete stent contact to the vessel wall, and edge
dissection; neoatherosclerosis plays a large role in very late The P2Y12 inhibitors exert their antiplatelet effects by inhi-
ST (Adriaenssens 2014). It is suggested that after DES place- bition of platelet ADP receptors. The inhibition prevents acti-
ment, late and very late ST are caused by vascular damage vation of the GP IIb/IIIa receptor complex and subsequent
and impaired reendothelialization. platelet activation and aggregation. Table 2 summarizes
some of the landmark trials comparing ticagrelor, prasu-
In-Stent Restenosis grel, and intravenous cangrelor with clopidogrel in the set-
The most common cause of ISR after stent implantation is ting of ACS. Guideline recommendations have echoed those
the occurrence of severe neointimal proliferation—a phe- results, preferring ticagrelor and prasugrel over clopidogrel in
nomenon involving tunica intima hyperplasia of the coro- the treatment of patients with ACS (Collet 2021; Amsterdam
nary artery that ultimately results in thickening of the arterial 2014). Cangrelor can be a therapeutic option for patients who
wall and, thus, reduced lumen space (Dangas 2010). Patients have not been given a P2Y12 inhibitor before coronary angio-
with ISR typically present with recurrent angina, electro- gram and PCI (Collet 2021).
cardiogram changes related to ischemia, or abnormal inva- Most recently, the ISAR-REACT 5 trial sought to answer
sive functional diagnostic tests (i.e., fractional flow reserve, the long-standing question of prasugrel versus ticagrelor—
intravascular ultrasound). It is important to note that DAPT particularly in non-ST-elevation acute coronary syndrome
does not influence the development of ISR (Neumann 1996). (NSTE-ACS) (Schüpke 2019). This open-label, superiority trial

PSAP 2022 Book 1 • Cardiology 85 Antithrombotic Therapy in Cardiac Interventions


Table 2. Efficacy and Safety Comparison of P2Y12 Inhibitors

Trial (year) Patient population Intervention Outcomes Conclusion

TRITON-TIMI 38 Patients with Prasugrel 60-mg Primary Prasugrel demonstrated


(2007) moderate- to high- loading dose; then • Death from CV causes, a greater rate of life-
(prasugrel vs. risk ACS with 10 mg daily for nonfatal MI, or nonfatal threatening bleeding but
clopidogrel) scheduled PCI 6–15 months stroke: 9.9% vs. 12.1% was associated with lower
Vs. (p<0.001) rates of ischemic events
Clopidogrel 300- compared with clopidogrel
mg loading dose; Secondary
then 75 mg daily for • Major bleeding: 2.4% vs.
6–15 months 1.8% (p=0.03)
• Stent thrombosis: 1.1% vs.
2.4% (p<0.001)

PLATO (2009) Patients admitted Ticagrelor 180-mg Primary Ticagrelor was associated
(ticagrelor vs. with ACS, with loading dose followed • Composite of death from with lesser rates of
clopidogrel) or without by 90 mg twice daily vascular causes, MI, or ischemic events but was
ST-segment Vs. stroke at 12 months: 9.8% associated with more non-
elevation Clopidogrel 300– vs. 11.7% (p=0.001) procedure-related bleeding
600-mg loading dose
followed by 75 mg Secondary
daily • MI: 5.8% vs. 6.9%
(p=0.005)
• Death from vascular
causes: 4.0% vs. 5.1%
(p=0.001)
• Stroke: 1.5% vs. 1.3%
(p=0.22)
• Death from any cause:
4.5% vs. 5.9% (p=0.001)
• Major bleeding: 11.6% vs.
11.2% (p=0.43)
• Non-CABG-related major
bleeding: 4.5% vs. 3.8%
(p=0.03)

CHAMPION Patients with stable Cangrelor 30-mcg/kg Primary (cangrelor vs. Compared with clopidogrel,
PHOENIX or unstable CAD bolus followed by clopidogrel, respectively) cangrelor reduced early
(2016) undergoing PCI 4-mcg/kg/min bolus • Composite (death, ischemic events after PCI,
(cangrelor vs. for the duration of the MI, ischemia-driven including stent thrombosis
clopidogrel) PCI plus placebo pill revascularization, or stent
vs. clopidogrel 300– thrombosis at 48 hours):
600-mg loading dose 4.7% vs. 5.9% (p=0.005)
plus placebo bolus
or infusion Secondary
• Composite primary
outcome at 30 days:
6.0% vs. 7.0% (p=0.03)
• Stent thrombosis at
48 hours: 0.8% vs. 1.4%
(p=0.01)
• MI at 48 hours: 3.8% vs.
4.7% (p=0.02)
• Severe bleeding at
48 hours: 0.16% vs. 0.11%
(p=0.44)

(continued)

PSAP 2022 Book 1 • Cardiology 86 Antithrombotic Therapy in Cardiac Interventions


Table 2. Efficacy and Safety Comparison of P2Y12 Inhibitors  (continued)

Trial (year) Patient population Intervention Outcomes Conclusion

ISAR-REACT 5 Patients presenting Ticagrelor (180-mg Primary Adverse outcomes were


(2019) with ACS and loading dose followed • Composite (death, MI, lower in patients who
(ticagrelor vs. planned—or by 90 mg twice daily) stroke) at 1 year: 9.3% vs. received prasugrel
prasugrel) uncertain— Vs. 6.9% (p=0.006) compared with ticagrelor
invasive evaluation Prasugrel (60 mg- without a difference in
loading dose followed Secondary major bleeding
by 10 mg daily and • Bleeding (BARC 3–5):
adjusted to 5 mg daily 5.4% vs. 4.8% (p=0.48) This trial not only compared
per package insert) two antiplatelet drugs,
but also compared two
treatment strategies
(upstream vs. downstream
administration)

ACS = acute coronary syndrome; BARC = Bleeding Academic Research Consortium; CABG = coronary artery bypass grafting; CAD =
coronary artery disease; CV = cardiovascular; MI = myocardial infarction; PCI = percutaneous coronary intervention.
Information from: Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus clopidogrel in patients with acute coronary
syndromes. N Engl J Med 2007;357:2001-15; Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus clopidogrel in patients with
acute coronary syndromes. N Engl J Med 2009;361:1045-57; Schüpke S, Neumann F-J, Menichelli M, et al. Ticagrelor or prasugrel in
patients with acute coronary syndromes. N Engl J Med 2019;381:1524-34; Abtan J, Steg PG, Stone GW, et al. Efficacy and safety of
cangrelor in preventing periprocedural complications in patients with stable angina and acute coronary syndromes undergoing
percutaneous coronary intervention: the CHAMPION PHOENIX trial. JACC Cardiovasc Interv 2016;9:1905-13.

randomized more than 4000 patients to receive either tica- Upstream vs. Downstream Administration
grelor or prasugrel. In the ticagrelor arm, a loading dose was In the setting of NSTE-ACS, the optimal timing of oral P2Y12
administered as soon as possible after randomization. For inhibitor administration remains elusive. Administration of
those randomized to prasugrel, the loading dose was delayed an oral P2Y12 inhibitor before coronary angiography when
in patients with NSTE-ACS undergoing PCI, which was per- the coronary anatomy is unknown—also called pretreat-
formed in a majority (84.1%) of patients, 58.9% of whom had ment or upstream strategy, detailed in Figure 1—may, theo-
NSTE-ACS. The primary endpoint was a composite of death, retically, reduce the risk of ischemia while patients wait to
myocardial infarction (MI), and stroke and occurred in 6.3% undergo coronary angiography (Tarantini 2020). In addition,
of patients randomized to prasugrel compared with 9.3% of the achievement of sufficient platelet inhibition at the time
patients randomized to ticagrelor (HR 1.36; 95% CI, 1.09– of PCI may reduce the risk of periprocedural thrombotic com-
1.70; p=0.006). This was driven primarily by a reduction in MI plications caused by vascular damage and impaired reendo-
(3.7% taking prasugrel vs. 4.8% taking ticagrelor; HR 1.63; 95% thelialization of the coronary arteries. However, an upstream
CI, 1.18–2.25). Surprisingly, there were no significant differ- strategy may also increase the risk of periprocedural bleeding
ences in rates of major bleeding between prasugrel and tica- during PCI or coronary artery bypass graft (CABG) and there-
grelor, but that could have been attributed to certain design fore increase hospital length of stay and cost.
limitations such as telephone follow-up for most patients, Table 3 lists current guideline recommendations for the
differences in pretreatment, lack of formal assessment of use of P2Y12 inhibitors with regard to timing of PCI. In the set-
medication adherence, and the exclusion of more patients in ting of SIHD, the recommendation is to administer a loading
the safety analysis in the prasugrel arm than the ticagrelor dose of a P2Y12 inhibitor before PCI once coronary anatomy
arm. But regardless of those limitations, this trial resulted in is known (e.g., coronary angiogram is performed) and once
the ESC’s inclusion of a class IIb guideline recommendation the decision has been made to perform an intervention (Neu-
favoring prasugrel over ticagrelor in the treatment of patients mann 2019). In the setting of ST-segment-elevation myocar-
with NSTE-ACS going for PCI (Collet 2021). Ultimately, the dial infarction (STEMI), P2Y12 administration should occur
selection of a P2Y12 inhibitor should consider several charac- as soon as possible—or at least at the time of PCI (Neumann
teristics, including clinical presentation (e.g., ACS vs. SIHD), 2019). In NSTE-ACS, upstream administration (or pretreat-
ischemic and bleeding risk factors, and cost. Further stud- ment) may be considered for clopidogrel and ticagrelor, but
ies are warranted to confirm the results demonstrated by guidelines recommend against upstream administration of
ISAR-REACT 5. prasugrel based on results of the ACCOAST trial detailed later.

PSAP 2022 Book 1 • Cardiology 87 Antithrombotic Therapy in Cardiac Interventions


Upstream Strategy P2Y12 Inhibitor Loading Dose Coronary Angiography PCI

NSTE-ACS
PCI P2Y12 Inhibitor
Downstream Strategy Coronary Angiography +/– P2Y12 Inhibitor at start of (if not given at time of PCI)
PCI

Figure 1. Upstream vs. downstream P2Y12 administration relative to timing of PCI in NSTE-ACS.
ACS = acute coronary syndrome; NSTE = non-ST elevation; PCI = percutaneous coronary intervention.

Table 3. Clinical Guideline Recommendations Regarding Timing of P2Y12 Administration

Recommendations

2013 ACC/AHA STEMI guidelines A P2Y12 inhibitor loading dose should be given as early as possible or at time of PCI
(class Ib)
2014 AHA/ACC NSTE-ACS guidelines A P2Y12 inhibitor loading dose should be given before PCI with stenting (class Ia)
2018 ESC/EACTS myocardial SIHD
interventional guidelines Pretreatment with clopidogrel is recommended in patients undergoing elective PCI
once the coronary anatomy is known and a decision has been made to proceed with
PCI (class Ia)

NSTE-ACS
• Ticagrelor may be administered before PCI, and clopidogrel may be used if ticagrelor
is not available (class IIa)
• Administration of prasugrel in patients with unknown coronary anatomy is not
recommended (class III)

STEMI
Prasugrel or ticagrelor (clopidogrel if prasugrel or ticagrelor is either not available or
contraindicated) is recommended before or at least at the time of PCI (class Ia)
2020 ESC ACS without STEMI NSTE-ACS
guidelines • Pretreatment may be considered in patients who are not planning to undergo an
early invasive strategy (i.e., coronary angiography within 24–72 hours) and who do
not have high bleeding risk (class IIb)
• Not recommended to administer routine pretreatment P2Y12 when coronary anatomy
is not known and an early invasive management is planned (class III)

ACC = American College of Cardiology; ACS = acute coronary syndrome; AHA = American Heart Association; CAD = coronary artery
disease; ESC = European Society of Cardiology; NSTE-ACS = non-ST-elevation acute coronary syndrome; PCI = percutaneous coronary
intervention; SIHD = stable ischemic heart disease; STEMI = ST-segment-elevation myocardial infarction.
Information from: Amsterdam EA, Wenger NK, Brindis RG, et al. 2014 ACC/AHA guideline for the management of patients with
non-ST-elevation acute coronary syndromes: a report of the American College of Cardiology/American Heart Association Task
Force on Practice Guidelines. Circulation 2014;130:e344-e426; O’Gara PT, Kushner FG, Ascheim DD, et al. 2013 ACCF/AHA guideline
for the management of ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American
Heart Association Task Force on Practice Guidelines. Circulation 2013;127;e362-425; Neumann F-J, Sousa-Uva M, Ahlsson A, et al.
2018 ESC/EACTS guidelines on myocardial revascularization. Eur Heart J 2019;40:87-165. Collet J-P, Thiele H, Barbato E, et al. 2020
ESC guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation.
Eur Heart J 2021;42:1289-367.

Literature Review Evaluating Timing angiography (upstream) compared with known coronary angi-
of P2Y12 Administration ography status—if PCI was indicated (downstream). Patients
ACCOAST in the upstream arm received a prasugrel 30-mg loading
The ACCOAST trial evaluated the effect of prasugrel adminis- dose before coronary angiography, and patients in the down-
tration at the time of NSTE-ACS diagnosis and before coronary stream arm received placebo. After the decision to undergo

PSAP 2022 Book 1 • Cardiology 88 Antithrombotic Therapy in Cardiac Interventions


PCI, an additional 30 mg of prasugrel was administered to 2016). Morphine can slow gastric emptying, thereby slow-
the upstream group, and 60 mg to the downstream group. In ing absorption of oral P2Y12 inhibitors, potentially diminish-
this study, the use of an upstream prasugrel loading strategy ing the desired rapid platelet inhibition, and thus increasing
did not prove to be more efficacious in reducing major isch- the risk of ischemic events. Several trials have demonstrated
emic events compared with the downstream strategy and reduced antiplatelet effects, including delayed maximal P2Y12
was associated with increased risk of major bleeding events inhibitor plasma concentrations, reduced overall P2Y12 inhibi-
(Montalescot 2013). As a result of this trial, both the American tor exposure, and higher rates of ischemic events associated
and European guidelines recommend against the upstream with opioid administration (Table 4).
administration of prasugrel in NSTE-ACS (Class III: HARM).
Fentanyl for Procedural Sedation in PCI
DUBIUS Similarly, fentanyl is routinely utilized for procedural seda-
The DUBIUS trial compared downstream versus upstream oral tion in patients undergoing PCI. The PACIFY trial (see Table 4)
P2Y12 inhibitor administration strategies in patients with NSTE- described the impact of fentanyl on ticagrelor plasma concen-
ACS undergoing invasive treatment (i.e., coronary angiography trations and its antiplatelet effect. The trial demonstrated a
within 24–72 hours of presentation). Patients in the upstream reduction in ticagrelor plasma concentrations, higher platelet
administration arm received a ticagrelor loading dose before reactivity tested by VerifyNow and platelet aggregometry, and
angiography, and those in the downstream administration arm an increase in troponin-I levels postprocedure. VerifyNow is an
received no P2Y12 inhibitor before angiography. After angi- available point-of-care blood test that measures responsive-
ography, patients in the downstream arm were then random- ness to P2Y12 receptor blockade, measured by platelet reactive
ized to receive a ticagrelor or prasugrel loading dose before units. A result below the threshold cutoff indicates an antiplate-
PCI. Upon interim analysis, the trial was stopped prematurely let effect; values above the cutoff indicate a lack of antiplate-
because of futility. Neither the rate of the primary end point, let effect. In addition to those findings, self-reported maximal
death due to vascular causes, nonfatal MI, nonfatal stroke, nor intraprocedural pain was low at a median of 1.5 on a 10-point
Bleeding Academic Research Consortium (BARC) types 2, 3, scale, further supporting the need to reconsider the routine use
and 4 bleeding at 30 days differed between the downstream of fentanyl in patients undergoing PCI (McEvoy 2018).
and upstream groups, respectively (2.9% and 3.3%; ARR –0.46;
Practical Solutions and Considerations
95% CI, –2.87 to 1.89). Given the minimal difference in out-
comes between the groups, it was deemed unlikely that one Upon the results of these clinical trials (see Table 4), the Food
strategy was preferred over the other (Tarantini 2020). and Drug Administration updated the prescribing information
for clopidogrel, prasugrel, and ticagrelor in 2018 to include
Practical Considerations a warning about the interaction with opioids and to recom-
In the setting of NSTE-ACS, pretreatment with clopidogrel and mend consideration of a parenteral antiplatelet (cangrelor or
ticagrelor should be patient specific depending on ischemic GP IIb/IIIa inhibitors) agent in patients with ACS who require
and bleeding risk factors. The GRACE score has been found opioid coadministration (Furtado 2020). In clinical practice,
to be a possible tool for determining patients who may require it is important to discourage the routine use of opioids in the
CABG and thus may benefit from deferring upstream admin- absence of refractory chest pain—particularly in close prox-
istration, but it has not been fully validated in clinical trials imity to oral P2Y12 inhibitor administration. There are cur-
(Avci 2015). However, in patients who are ultimately deemed rently no recommendations to routinely assess impaired
unsuitable for CABG and who undergo PCI as the revascu- P2Y12 inhibitor plasma concentrations or heightened platelet
larization strategy after coronary anatomy is known, admin- reactivity with various testing modalities in patients receiv-
istration of a P2Y12 inhibitor as soon as possible before PCI ing opioids, but such a testing option may be considered
is reasonable. Because of the harm noted in the ACCOAST in selected cases (e.g., increased bleeding risk, increased
study, prasugrel should not be given routinely before angiog- thrombotic risk, planned escalation or de-escalation of anti-
raphy. For patients with STEMI, it is reasonable to administer platelet therapy) (Sibbing 2019). It is interesting that meto-
P2Y12 inhibitors once diagnosis has been made (O’Gara 2013). clopramide administered concomitantly with morphine and
ticagrelor demonstrated improvements in antiplatelet activ-
Interaction Between Opioids and P2Y12 ity (Saad 2020). There may be a role for prokinetic agents
Inhibitors such as metoclopramide in this setting, though more stud-
Morphine in Acute Coronary Syndromes ies are needed to determine the effect on clinical outcomes.
Morphine is often used for chest pain associated with ACS
(Amsterdam 2014, O’Gara 2013). Although morphine pro- DAPT Duration After PCI

vides patients with symptomatic relief, the pharmacologic Clinical Practice Guideline Recommendations
interaction between opioids and a blunted effect of P2Y12 Recommendations from the most-recent ACC/AHA Guide-
inhibitors has become of concern (Furtado 2020; Kubica lines for duration of DAPT after PCI are given in Table 5

PSAP 2022 Book 1 • Cardiology 89 Antithrombotic Therapy in Cardiac Interventions


Table 4. Interaction Between Opioids and P2Y12 Inhibitors

Trial (year) Patient Population Intervention Outcomes Conclusion

IMPRESSION (2016) Patients with acute Morphine 5 mg IV Primary Morphine exerted


MI vs. placebo • Ticagrelor AUC(0–12h): 36% a negative impact
followed by decrease (p=0.003) on ticagrelor by
ticagrelor 180 mg lowering total
Secondary exposure to
• Ticagrelor AUC(0–6h): 55% decrease ticagrelor and its
(p=0.002) active metabolite
• Ticagrelor active metabolite by 36% and delaying
AUC(0–6h): 53% decrease (p=0.006) its maximal plasma
• Ticagrelor Tmax: 2-hour delay concentration by
(p=0.004) 2 hours
• Ticagrelor Cmax: 1156 ± 771 vs.
1683 ± 847 ng/mL (p=0.006)

PACIFY (2018) Patients Fentanyl vs. Primary (fentanyl vs. no fentanyl, Fentanyl
undergoing no fentanyl respectively) administration
elective coronary for procedural • Ticagrelor AUC(0–24h): 2107 vng lowers plasma
angiography and sedation ml–1 h vs. 3301 vng ml–1 h (p=0.05) concentrations of
receiving ticagrelor ticagrelor and delays
Secondary (fentanyl vs. no fentanyl, its antiplatelet
respectively) effects
• 2-hour mean ± SD P2Y12 reaction
unit value: 112 ± 95 vs. 78 ± 72
(p=0.09)
• ADP response by aggregometry:
39.3 ± 8.7% vs. 27.5 ± 14.4%
(p=0.04)
• P2Y12 reaction unit values at
4 hours: 55 vs. 50 (p=0.73)
• Mean self-reported maximal
intraprocedural pain (10-point
numeric scale): 1.5 vs. 2.3
(p=0.14)

ATLANTIC- Patients with Morphine Primary (morphine vs. no morphine, The use of morphine
Morphine (2019) STEMI undergoing Vs. respectively) was associated with
PCI in the No morphine + Coprimary endpoint: increased GP IIb/IIIa
ATLANTIC trial ticagrelor 180 mg • Absence of pre-PCI TIMI 3 flow in inhibitor use, less
followed by 90 mg culprit artery: OR 1.54 (1.19, 1.99) pre-PCI TIMI 3 flow,
twice daily (p=0.001) and more bleeding
• Absence of pre-PCI ≥70%
ST-segment elevation resolution:
OR 1.32 (0.98, 1.77) (p=0.07)

Clinical end points (morphine vs. no


morphine, respectively)
• Absence of pre-PCI TIMI 3 flow:
85.8% vs. 79.7% (p=0.001)
• TIMI major bleeding: 1.1% vs. 0.1%
(p=0.02)
• Bailout use of GP IIb/IIIa
inhibitors: 11.3% vs. 7.9% (p=0.01)

(continued)

PSAP 2022 Book 1 • Cardiology 90 Antithrombotic Therapy in Cardiac Interventions


Table 4. Interaction Between Opioids and P2Y12 Inhibitors (continued)

Trial (year) Patient Population Intervention Outcomes Conclusion

EARLY ACS Patients treated Patients treated Outcomes The use of morphine
subanalysis (2020) with clopidogrel with clopidogrel • Composite of death, MI, recurrent in combination with
presenting with within 24 hours ischemia, or thrombotic bailout clopidogrel was
non-ST-segment of randomization at 96 hours (four-way end point): associated with
elevation ACS in Vs. adjusted OR 1.40 (95% CI, 1.04– higher rates
the EARLY ACS Patients not 1.87) (p=0.026) of ischemic events
trial treated with • Composite of death or MI at 30
clopidogrel days: adjusted OR 1.29 (95% CI,
(negative control) 0.98–1.70) (p=0.072)
Both groups
received
morphine
prerandomization

ACS = acute coronary syndrome; ADP = adenosine diphosphate; AUC = area under the curve; CI = confidence interval; IV = intrave-
nously; MACE = major adverse cardiovascular events; MI = myocardial infarction; OR = odds ratio; PCI = percutaneous coronary inter-
vention; SD = standard deviation; STEMI = ST-segment-elevation myocardial infarction; TIMI = thrombolysis in myocardial
infarction.
Information from: Kubica J, Adamski P, Ostrowska M, et al. Morphine delays and attenuates ticagrelor exposure and action in
patients with myocardial infarction: the randomized, double-blind, placebo-controlled IMPRESSION trial. Eur Heart J 2016;37:245-
52; McEvoy JW, Ibrahim K, Kickler TS, et al. Effect of intravenous fentanyl on ticagrelor absorption and platelet inhibition among
patients undergoing percutaneous coronary intervention: the PACIFY randomized clinical trial (platelet aggregation with ticagrelor
inhibition and fentanyl). Circulation 2018;137:307-9; Lapostolle F, Van’t Hof AW, Hamm CW, et al. Morphine and ticagrelor interaction
in primary percutaneous coronary intervention in ST-segment elevation myocardial infarction: ATLANTIC-Morphine. Am J
Cardiovasc Drugs 2019;19:173-83; Montalescot G, van ’t Hof AW, Lapostolle F, et al. Prehospital ticagrelor in ST-segment elevation
myocardial infarction. N Engl J Med 2014;371:1016-27; Furtado RHM, Nicolau JC, Guo J, et al. Morphine and cardiovascular
outcomes among patients with non-ST-segment elevation acute coronary syndromes undergoing coronary angiography. J Am Coll
Cardiol 2020;75:289-300.

Table 5. ACC/AHA Guidelines for DAPT Duration After PCI

Recommendations High Bleed Risk Low Bleed Risk

SIHD BMS: 1 month DES: 3 months (Class IIb) BMS: >1 month if high thrombotic
DES: 6 months BMS: 1 month riska
Clopidogrel preferred DES: >6 months if high
(Class I) thrombotic risk (Class IIb)

ACS 12 months 6 months regardless of stent >12 months regardless of stent


Ticagrelor or prasugrel type (Class IIb) type
preferred (Class IIa) No preferred P2Y12 inhibitor
(Class IIb)

a
Advanced age, multiple previous myocardial infarctions, extensive coronary artery disease, diabetes mellitus, chronic kidney
disease
ACS = acute coronary syndrome; BMS = bare-metal stent; DES = drug-eluting stent; SIHD = stable ischemic heart disease.
Information from: Levine GN, Bates ER, Bittl JA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet
therapy in patients with coronary artery disease. Circulation 2016;134:e123-55.

PSAP 2022 Book 1 • Cardiology 91 Antithrombotic Therapy in Cardiac Interventions


(Levine 2016). In general, after PCI with a DES, DAPT is Shorter DAPT Durations
recommended for at least 6 and 12 months for SIHD and Since publication of the AHA/ACC DAPT guidelines, several
ACS, respectively. Shorter durations can be considered for clinical trials have evaluated shorter durations of DAPT after
patients at low ischemic risk and high bleeding risk, whereas PCI with stent placement—in the settings of both SIHD and
longer durations can be considered for those with high isch- ACS. Those trials are summarized in Table 6. Overall, the
emic and low bleeding risks. regimens and durations of DAPT followed by P2Y12 inhibitor

Table 6. Literature Review: Evaluating Shorter DAPT Durations

Trial (year)a Patient Population Intervention Outcomes Conclusion

GLOBAL Patients DAPT for Primary Ticagrelor


LEADERS (2018) undergoing PCI 1 month All-cause mortality or nonfatal MI: monotherapy for
with DES followed by 1 month DAPT 3.8% vs. 12 months 23 months after
ACS 50% ticagrelor for DAPT 4.4% (p=0.073) 1 month of DAPT
CAD 50% 23 months failed to show
Vs. Secondary (1 month DAPT followed by superiority to
DAPT for ticagrelor vs. 12 months DAPT followed standard therapy
12 months by aspirin, respectively)
followed by • All-cause mortality: 2.8% vs. 3.2%
aspirin for (p=0.18)
12 months • MI: 1.0% vs. 1.3% (p=0.14)
• Grade 3 or 5 BARC bleeding: 2.0% vs.
2.1% (p=0.77)

TWILIGHT (2019) Patients Aspirin + Primary Shorter DAPT duration


undergoing PCI ticagrelor for BARC 2, 3, or 5 bleeding at 12 months: resulted in less
(DES) with ≥1 3 months and ticagrelor monotherapy 4.0% vs. bleeding
high-risk feature then ticagrelor ticagrelor + aspirin 7.1% (p=0.001)
of ischemia or monotherapy
bleeding Vs. Secondary
ACS 65% Ticagrelor + All-cause mortality, MI, stroke: 3.9% vs.
CAD 35% aspirin for 12 3.9% (p for noninferiority = 0.001)
months MI 2.7% vs. 2.7% (p=0.001);
stent thrombosis: 0.4% vs. 0.6%
(p>0.05);
ischemic stroke: 0.5% vs. 0.2% (p>0.05)

STOPDAPT-2 3000 Japanese 1 month of DAPT Primary With regard to adverse


(2019) patients (with either Death, MI, stent thrombosis, stroke, ischemic events,
undergoing PCI clopidogrel TIMI major/minor bleeding at 1 year: 1 month of DAPT was
with DES or prasugrel) 1 month DAPT 2.4% vs. 12 months superior to 12 months
ACS 62% followed by DAPT 3.7% (p for superiority = 0.04) of DAPT
CAD 38% clopidogrel
Mostly patients monotherapy Secondary (1 month vs. 12 months
with low to for up to 5 years DAPT, respectively)
intermediate Vs. • Death, MI, stent thrombosis,
ischemic risk 12 months of or stroke at 1 year: 2.0% vs. 2.5%
DAPT followed (p for noninferiority = 0.005)
by aspirin • TIMI major/minor bleeding at 1 year:
monotherapy 0.4% vs. 1.5% (p for superiority =
for 5 years 0.004)
• BARC 3 or 5 bleeding at 1 year: 0.5%
vs. 1.8% (p for superiority = 0.003)
• Definite or probable stent
thrombosis: 0.3% vs. 0.07% (p for
superiority = 0.21)

(continued)

PSAP 2022 Book 1 • Cardiology 92 Antithrombotic Therapy in Cardiac Interventions


Table 6. Literature Review: Evaluating Shorter DAPT Durations  (continued)

Trial (year)a Patient Population Intervention Outcomes Conclusion

SMART CHOICE Patients Aspirin + P2Y12 Primary After 3 months


(2019) undergoing PCI for 3 months Major adverse cardiac and of DAPT, P2Y12
with DES followed cerebrovascular events (a composite of monotherapy was
by P2Y12 all-cause death, myocardial infarction, noninferior compared
monotherapy or stroke) at 12 months: monotherapy with prolonged DAPT
vs. DAPT for 2.9% vs. DAPT 2.5% (p=0.007 for
12 months noninferiority)

Secondary (monotherapy vs. DAPT,


respectively)
• All-cause death: 1.4% vs. 1.2%
(p=0.61)
• MI: 0.8% vs. 1.2% (p=0.28
• Stroke: 0.8% vs. 0.3% (p=0.14)
• BARC bleeding 2–5: 2.0% vs. 3.4%
(p=.02)

TICO (2020) Patients Ticagrelor Primary Ticagrelor


undergoing monotherapy Death, MI, stent thrombosis, stroke, monotherapy after
PCI following after 3 months target vessel revascularization or TIMI 3 months of DAPT
after ACS with a of DAPT major bleeding at 12 months: ticagrelor was superior to
biodegradable DES vs. monotherapy after 3 months of DAPT standard therapy at
Standard DAPT 3.9% vs. standard DAPT therapy 5.9% 12 months
therapy for (p=0.01)
12 months
Secondary
Major bleeding: ticagrelor monotherapy
after 3 months of DAPT 1.7% vs.
standard DAPT therapy 3.0% (p=0.02)
Stent thrombosis: ticagrelor
monotherapy after 3 months of DAPT
0.4% vs. standard DAPT therapy 0.3%
(p=0.53)

a
All studies excluded patients who required chronic oral anticoagulation.
ACS = acute coronary syndromes; BARC = Bleeding Academic Research Consortium; CAD = coronary artery disease; DAPT = dual
antiplatelet therapy; DES = drug-eluting stent; MI = myocardial infarction; PCI = percutaneous coronary intervention; TIMI =
thrombolysis in myocardial infarction.
Information from: Vranckx P, Valgimigli M, Jüni P, et al. Ticagrelor plus aspirin for 1 month, followed by ticagrelor monotherapy for
23 months vs aspirin plus clopidogrel or ticagrelor for 12 months, followed by aspirin monotherapy for 12 months after implantation
of a drug-eluting stent: a multicentre, open-label, randomised superiority trial. Lancet 2018;392:940-9; Mehran R, Baber U, Sharma
SK, et al. Ticagrelor with or without aspirin in high-risk patients after PCI. N Engl J Med 2019;381:2032-42; Watanabe H, Domei T,
Morimoto T, et al. Effect of 1-month dual antiplatelet therapy followed by clopidogrel vs 12-month dual antiplatelet therapy on
cardiovascular and bleeding events in patients receiving PCI: the STOPDAPT-2 randomized clinical trial. JAMA 2019;321:2414; Hahn
J-Y, Song YB, Oh J-H, et al. Effect of P2Y12 inhibitor monotherapy vs dual antiplatelet therapy on cardiovascular events in patients
undergoing percutaneous coronary intervention: the SMART-CHOICE randomized clinical trial. JAMA 2019;321:2428; Kim B-K, Hong
S-J, Cho Y-H, et al. Effect of ticagrelor monotherapy vs ticagrelor with aspirin on major bleeding and cardiovascular events in
patients with acute coronary syndrome: the TICO randomized clinical trial. JAMA 2020;323:2407.

monotherapy varied widely from trial to trial. DAPT duration In general, the findings from the studies demonstrated com-
ranged from 1 to 3 months post-PCI followed by P2Y12 inhibi- parable ischemic efficacy while reducing bleeding events
tor monotherapy for up to 12–24 months. The regimens were with P2Y12 inhibitor monotherapy compared with DAPT.
compared with standard of care, which consists of DAPT Although shortened DAPT duration has not become incor-
for 6–12 months post-PCI followed by aspirin monotherapy. porated into clinical practice guidelines as yet, it is reasonable

PSAP 2022 Book 1 • Cardiology 93 Antithrombotic Therapy in Cardiac Interventions


for clinicians to consider shortening DAPT duration—particu-
Box 1. Clinical Characteristics of
larly in patients with higher bleeding risks or histories of pre-
Patients Who May Benefit from
vious bleeding while on DAPT. However, in the application of
Extended-DAPT Duration
these regimens in clinical practice, it is imperative that stent
ACS presentation/previous ACS event
type be considered. Although some of this evidence is in the
Current cigarette use
setting of DES placement, some trials have used bioresorb- Diabetes mellitus
able stents. Therefore, appropriate application of these trials Heart failure (both reduced and preserved ejection
will have to be done with the type of stent in mind. fraction)
High CAD burden
Extended-DAPT Duration Increased procedure complexity
On the other hand, some evidence suggests that longer Left ventricular ejection fraction < 30%
Older-generation stent(s)
courses of DAPT—beyond 12 months—could be beneficial.
Peripheral arterial disease
Extended-DAPT duration can be considered after PCI for up Renal dysfunction
to 30 months. But even though that approach has demon- Stent diameter < 3 mm
strated reduced risk of myocardial infarction, it comes with Vein graft PCI
an increased risk of bleeding. Because of that, patients who
ACS = acute coronary syndrome; CAD = coronary artery dis-
may be most optimal for this strategy are somewhat difficult
ease; DAPT = dual antiplatelet therapy; PCI = percutaneous
to identify. Patients with the high-risk characteristics listed coronary intervention.
in Box 1 may potentially benefit from extended-DAPT dura- Information from: Howard CE, Nambi V, Jneid H, et al. Extended
tion. But it is important to note that the promising evidence of duration of dual-antiplatelet therapy after percutaneous cor-
onary intervention: how long is too long? J Am Heart Assoc
extended-DAPT therapy is derived from studies in which old- 2019;8:e012639.
er-generation stents were used (Howard 2019). Such stents
pose a higher risk of late in-stent thrombosis, and so, the ben-
efits of extended DAPT in the placement of newer stents are bleeding risk factors, which can be assessed by using the
unknown. Ultimately, the approach to DAPT duration after PCI DAPT score and PRECISE-DAPT score as described in Table 7
should be individualized based on a patient’s ischemic and and Table 8, respectively.

Table 7. DAPT Score

Points Interpretation

Age (years) Score ≥2:


75 –2 Favorable risk–benefit ratio for prolonged
65–74 –1 DAPT (>12 months)
<65 0

Cigarette smokinga +1

Diabetes mellitus +1

MI at presentation +1

Prior PCI or previous MI +1 Score <2:


Unfavorable benefit/risk ratio for prolonged
Paclitaxel-eluting stent +1
DAPT
Stent diameter <3 mm +1

LVEF <30% +2

Vein graft stent +2

a
Smoking within 1 year before index procedure.
DAPT = dual antiplatelet therapy; LVEF = left ventricular ejection fraction; MI = myocardial infarction; PCI = percutaneous coronary
intervention.
Information from: Yeh RW, Secemsky EA, Kereiakes DJ, et al. Development and validation of a prediction rule for benefit and harm of
dual antiplatelet therapy beyond 1 year after percutaneous coronary intervention. JAMA 2016;315:1735; Levine GN, Bittl JA, Brindis
RG, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease.
J Am Coll Cardiol 2016;68:1082-115.

PSAP 2022 Book 1 • Cardiology 94 Antithrombotic Therapy in Cardiac Interventions


Table 8. PRECISE-DAPT Score

Points Interpretation

Hemoglobin (g/dL) 0–15 Total score ≥ 25


High PRECISE-DAPT score
Age (years) 0–19
Shortened duration of DAPT (3–6 months)
White blood cells (10 units/mcL)
3
0–15 should be considered

Creatinine clearance (mL/min) 0–25 Total score < 25


Nonhigh PRECISE DAPT score
Prior major bleeding 0–26
Longer durations of DAPT (12–24 months)
should be considered

Total score 0–100


To calculate score, use the online calculator at http://precisedaptscore.com/predapt/index.html

Information from: Costa F, van Klaveren D, James S, et al. Derivation and validation of the predicting bleeding complications in patients
undergoing stent implantation and subsequent dual antiplatelet therapy (PRECISE-DAPT) score: a pooled analysis of individual-
patient datasets from clinical trials. Lancet 2017;389:1025-34.

Switching Between Different P2Y12 Inhibitors based on the pharmacodynamic properties of the agents and
Switching Between Oral P2Y12 Inhibitors previously published literature (Figure 2).
For several reasons—including adverse effects, cost, and
other clinical scenarios—switching between oral P2Y12 inhib- SWAP-4 Study
itors is common in clinical practice. There are two scenarios The SWAP-4 study was a prospective, randomized, open-
in the switching of P2Y12 inhibitors: de-escalation and escala- label, single-center study to assess the timing, dosing, and
tion. De-escalation involves transitioning from a more-potent impact of the commonly occurring need to switch P2Y12
P2Y12 inhibitor to a less-potent option (e.g., ticagrelor or pras- classes in clinical practice in a de-escalation from tica-
ugrel to clopidogrel). Escalation involves transitioning from a grelor to clopidogrel therapy. The study was conducted in
less-potent P2Y12 inhibitor to a more-potent option (e.g., clopi- 80 patients receiving maintenance doses of aspirin and clopi-
dogrel to ticagrelor or prasugrel). Switching from ticagrelor dogrel for at least 30 days. After a 7-day run- in period of tica-
to prasugrel or vice versa is simply referred to as a change grelor (180 mg loading dose followed by 90 mg twice daily),
in therapy. Several considerations must be considered before patients were randomized to one of four groups and contin-
a switch in agents, including timing of the index event (i.e., ued for 7–13 days, in addition to aspirin:
date of stent placement), potential drug-drug interactions
when agents overlap, and potential loading-dose require- • Group A: Clopidogrel 600 mg administered 24 hours after
the last dose of ticagrelor
ments. A drug-drug interaction is not expected to occur in
a switch of agents within the same class (e.g., clopidogrel • Group B: Clopidogrel 600 mg administered 12 hours after
the last dose of ticagrelor
to prasugrel), but a drug-drug interaction is likely to occur
in a switch between classes (e.g., ticagrelor to clopidogrel). • Group C: Clopidogrel 75 mg daily administered 24 hours
after the last dose of ticagrelor
Because of the fast offset (3–5 days) and reversible receptor-
binding properties of ticagrelor, de-escalation to clopidogrel
• Group D: Ticagrelor 90 mg twice daily
produces a gap in platelet inhibition upon ticagrelor discon- Pharmacodynamic assessments of platelet reactiv-
tinuation, which could increase the risk of stent thrombosis. ity were performed at baseline with three platelet reactivity
Therefore, a loading dose of clopidogrel is generally recom- tools—after the run-in phase, and at 2, 24, 48, and 72 hours as
mended regardless of date of stent placement. With prasu- well as 10 days post randomization. Genetic status of the CYP
grel, there is still the potential for a lapse in platelet inhibition 2C19 enzyme was also tested in all patients at baseline. After
in a de-escalation to clopidogrel, but that is because of pras- a switch from clopidogrel to ticagrelor (run-in phase), platelet
ugrel’s irreversible platelet inhibition and longer offset. New reactivity by using three pharmacodynamic assays was sig-
platelets must be produced for clopidogrel to exert its anti- nificantly reduced compared with baseline levels (p<0.001).
platelet properties, so a loading dose of clopidogrel is still Compared with all clopidogrel groups, group D had lower plate-
recommended within 30 days of the index event in this set- let reactivity at all time points. Platelet reactivity remained
ting. Experts have produced a consensus recommendation significantly lower (p<0.001) in patients randomized to group

PSAP 2022 Book 1 • Cardiology 95 Antithrombotic Therapy in Cardiac Interventions


Figure 2. Recommendations on switching between oral P2Y12 inhibitors in acute and late phases.
Information from: Angiolillo DJ, Rollini F, Storey RF, et al. International expert consensus on switching platelet P2Y12 receptor–
inhibiting therapies. Circulation 2017;136:1955-75.

D compared with group A, B, or C and was similar between in patients undergoing PCI. It is also of note that CYP2C19
group A and group C (p=0.29). In group C, platelet reactiv- genetic status did not appear to have any meaningful impact
ity increased compared with baseline as early as 24 hours, on the study findings (Franchi 2018).
reaching statistical significance at 48 and 72 hours and up
to 10 days. De-escalation from ticagrelor therapy to clopido- TROPICAL-ACS
grel therapy was shown to be associated with an increase in With the use of potent P2Y12 inhibitors (ticagrelor, prasugrel),
platelet reactivity regardless of timing of ticagrelor discon- the greatest benefits are seen early after PCI, when the risk
tinuation and administration of a clopidogrel loading dose, of recurrent thrombotic events is highest. However, a higher
which was suggestive of a drug interaction that is consistent risk of bleeding is maintained during the chronic treatment
with the known pharmacodynamic properties of each drug phase. While maintaining prevention of thrombotic events in
described earlier. Therefore, consistent with previously pub- the early phase, efforts have been made to reduce the risk of
lished recommendations, results suggest that repeating a bleeding in the chronic phase through de-escalation of P2Y12
clopidogrel 600-mg loading dose may be required to facilitate inhibitor therapy. De-escalation is the process of switch-
a less-abrupt increase in platelet reactivity after de-escala- ing from a potent P2Y12 inhibitor to a less-potent agent (e.g.,
tion from ticagrelor and to achieve prompt, fully therapeutic clopidogrel) (Claassens 2020).
effects (Angiolillo 2017). However, barring any ticagrelor-re- The TROPICAL-ACS trial investigated the safety and effi-
lated adverse effects or cost restrictions, a strategy of de-es- cacy of early de-escalation from prasugrel to clopidogrel
calation cannot be routinely recommended and should be guided by platelet function testing. Patients who underwent
avoided early after an acute coronary event—especially PCI after an ACS event (STEMI or NSTEMI) were randomized

PSAP 2022 Book 1 • Cardiology 96 Antithrombotic Therapy in Cardiac Interventions


Table 9. Transition from Intravenous to Oral P2Y12 Inhibitor

TRANSITION TO

Clopidogrel Prasugrel Ticagrelor

Cangrelor 30 mcg/kg bolus 600-mg loading dose 60-mg loading dose 180-mg loading dose
and 4 mcg/kg/min infusion administered immediately administered immediately administered as soon as
after cangrelor after cangrelor discontinuation possible during cangrelor
discontinuation A loading dose may also be infusion or immediately after
administered 30 minutes cangrelor discontinuation
before the infusion is stoppeda

a
Cangrelor European Medicines Agency full prescribing information.
Information from: Angiolillo DJ, Rollini F, Storey RF, et al. International expert consensus on switching platelet P2Y12 receptor–
inhibiting therapies. Circulation 2017;136:1955-75.

to prasugrel for 2 weeks (control group) or prasugrel for and the coronary vessel stented. An alternative method that
7 days followed by clopidogrel for 7 days (guided de-escala- ensures appropriate cangrelor initiation timing is to perform
tion group). Platelet function testing was completed 2 weeks platelet function testing, but the means for such testing
after hospital discharge by using the Multiplate Analyzer, a may not be readily available at all institutions. In contrast,
rapid whole-blood assay that detects platelet function. After patients undergoing PCI who are receiving cangrelor will
platelet function testing, the control group remained on pra- require transition to an oral P2Y12 inhibitor after the proce-
sugrel for the remainder of the study (11.5 months). Low dure, and depending on which P2Y12 inhibitor is chosen for
responders, defined as having high platelet reactivity, were maintenance therapy, potential drug-drug interactions will
transitioned back to prasugrel; and good responders, defined drive transition instructions. Summarized consensus rec-
as not having high platelet reactivity, continued with clopido- ommendations for transitioning from intravenous to oral are
grel for the remainder of the study. With regard to the primary listed in Table 9 (Angiolillo 2017).
outcome of cardiovascular death, stroke, myocardial infarc-
tion, or BARC ≥2 bleeding, de-escalation of maintenance anti- Stable Ischemic Heart Disease Management
platelet therapy was noninferior to 12 months of prasugrel.
Antiplatelet Therapy for Secondary Prevention
De-escalation of P2Y12 inhibitors guided by platelet function
As part of the thrombotic response, platelet aggregation plays
testing is a viable option and may be considered for patients
a key role in plaque disruption, and therefore, antiplatelet
at elevated risk of bleeding (e.g., anemia, need for DAPT plus
agents are used for lessening the occurrence of these events.
oral anticoagulant therapy, clinically significant bleeding on
For secondary prevention of myocardial infarction and death,
current therapy, socioeconomic factors, adverse effects)
the 2012 ACC/AHA guidelines for the management of SIHD
(Claassens 2020). Current guidelines do not endorse a spe-
recommend treatment with aspirin 75–162 mg daily, contin-
cific platelet function test, and therefore, clinicians may be
ued indefinitely (class Ia). In patients with contraindication
limited to whatever test is available at their institutions (Sib-
to aspirin, clopidogrel is a reasonable alternative (class Ib).
bing 2017).
For certain patients who are at high risk, treatment with DAPT
Switching Between Oral and Intravenous (aspirin 75–162 mg plus clopidogrel 75 mg daily) may be con-
P2Y12 Inhibitors sidered (class IIb) (Fihn 2012). Similarly, the European Soci-
In a transition from an oral to an intravenous P2Y12 inhibitor, ety of Cardiology (ESC) recommends antithrombotic therapy
there is no lapse in platelet reactivity given the continuous with aspirin 75–100 mg daily in patients with previous myo-
inhibition achieved with intravenous therapy. Therefore, in cardial infarctions or revascularizations of chronic coronary
a transition from an oral agent to cangrelor, cangrelor 0.75 syndromes who are in sinus rhythm (class Ia), with an alterna-
mcg/kg/min infusion should be initiated within 72 hours of tive option of clopidogrel 75 mg daily for patients with aspirin
discontinuation of oral P2Y12 inhibitors (Angiolillo 2017). Cur- intolerance (class Ib) (Knuuti 2020).
rent recommendations suggest starting cangrelor within
3 or 4 days after discontinuation of prasugrel and within ASET Pilot Study
2 or 3 days after discontinuation of clopidogrel and ticagre- After recent studies evaluated shortened DAPT duration in
lor, and earlier initiation may be warranted depending on an effort to minimize bleeding risk while maintaining pro-
patient factors such as how recently the stent was placed tection from ischemia, the ASET study assessed the use

PSAP 2022 Book 1 • Cardiology 97 Antithrombotic Therapy in Cardiac Interventions


of prasugrel monotherapy after placement of an everolim- of 23 months (Eikelboom 2017). Based on results from that
us-eluting stent in 201 low-risk patients with stable CAD. The study, anticoagulation may be considered for secondary pre-
study was a single-arm, open-label trial with a stopping rule vention in patients who have stable atherosclerotic vascular
based on occurrence of stent thrombosis that would result in disease with elevated ischemic risk, as adopted by the 2019
patient recruitment termination. At the time of study enroll- and 2020 ESC guidelines.
ment, all patients were on standard DAPT therapy. At the
time of elective PCI for SIHD, aspirin was discontinued and
TRIPLE ANTITHROMBOTIC THERAPY
prasugrel was initiated and continued for 3 months. The pri-
mary ischemia end point was a composite of cardiac death, Introduction to Dual and Triple Antithrombotic
Therapy
spontaneous target vessel MI, or definite stent thrombosis.
The primary bleeding end point was BARC 3 and 5 bleeding About 30% of patients with atrial fibrillation (AF) also have
during 3 months of follow-up. Both the primary ischemic and coronary artery disease, and AF has been reported in 3% –
bleeding end points occurred in one patient, and no stent 12% of patients undergoing PCI (Saito 2019). It is important
thrombosis events occurred. Although the ASET study had to note that the antithrombotic strategies for AF and PCI are
several limitations, including small sample size, it may pave different from each other and therefore—because of differ-
the way for future, larger studies to assess this alternative ences in thrombus physiologies—are not interchangeable
antiplatelet therapy regimen after PCI in low-risk patients (You 2012). Oral anticoagulation (OAC) is the preferred anti-
with stable CAD—particularly those with high bleeding risk thrombotic therapy for stroke prevention in atrial fibrillation,
(Kogame 2020). whereas DAPT remains the standard of care for prevention of
major adverse cardiac events and stent thrombosis after PCI
Anticoagulation for Secondary Prevention (January 2019; Levine 2016).
In addition to low-dose aspirin, anticoagulation can be con- Triple antithrombotic therapy (TAT) was previously rec-
sidered for patients who are in sinus rhythm with moderate ognized as theoretically necessary and empirically recom-
to high ischemic risk and low bleeding risk (Knuuti 2020). And mended for patients with AF undergoing PCI (Saito 2019). The
therefore, to reduce the risk of MI, stroke, and cardiovascu- combination of OAC and DAPT is associated, unsurprisingly,
lar death, the 2019 ESC SIHD and 2020 ESC NSTE-ACS guide- with significant increases in major and minor bleeding. For
lines recommend the addition of low-dose rivaroxaban 2.5 mg example, the risk of nonfatal bleeding with TAT is four times
twice daily to aspirin in patients who are more than 1 year higher than with vitamin K antagonist (VKA) monotherapy,
post-MI or who have multivessel CAD. For patients with high and the risk of intracranial hemorrhage is twice that with
ischemic risk, this is labeled as a Class IIa recommendation DAPT (Saito 2019; Hansen 2010).
and as a Class IIb recommendation for those with moderate Dual antithrombotic therapy (DAT) refers to the combina-
risk. Currently, the 2014 ACC/AHA guidelines for the manage- tion of OAC plus a P2Y12 inhibitor, and it has emerged as a
ment of SIHD do not include anticoagulation as an option, but way to reduce bleeding events in patients without increasing
those guidelines were published before publications of clin- the risk of thromboembolic complications. Because the omis-
ical trials that evaluated anticoagulation for secondary pre- sion of OAC increases the risk of cardioembolic stroke and
vention (Collet 2021; Knuuti 2020). mortality in patients with AF and because the addition of a
P2Y12 inhibitor to aspirin reduces the risk of stent thrombosis
COMPASS after PCI, it was thought that the omission of aspirin might
The COMPASS trial was a double-blind study to assess the decrease bleeding without sacrificing efficacy against isch-
effectiveness of low-dose rivaroxaban 5 mg twice daily alone, emic events (Dewilde 2013; Connolly 2006). Since the first
rivaroxaban 2.5 mg twice daily plus aspirin, and aspirin alone publication of trials that evaluated DAT versus TAT in 2013,
for secondary prevention in patients with stable atheroscle- several published clinical trials have solidified DAT as the
rotic vascular disease. The primary outcome—a composite preferred antithrombotic strategy in most patients with AF
of cardiovascular death, stroke, or MI—occurred least in the undergoing PCI.
rivaroxaban 2.5 mg twice daily plus aspirin group, but more
major bleeding occurred. In a comparison of rivaroxaban plus Clinical Practice Guideline Recommendations
aspirin versus aspirin alone, the hazard ratio for the compos- The AHA, ACC, and ESC recently published updated state-
ite outcome was 0.76 (95% CI, 0.66–0.86; p<0.001). The haz- ments about TAT and DAT (Angiolillo 2021; Hindricks 2021;
ard ratio for rivaroxaban alone versus aspirin alone was 0.90 Kumbhani 2021) (Figure 3). In general, all favor DAT in most
(95% CI, 0.79–1.03; p=0.12). Recurrent cardiovascular events patients with AF undergoing PCI. For patients during the peri-
did not occur less often in the rivaroxaban 5 mg twice daily PCI period and during a hospitalization of up to 1 week, TAT is
group compared with aspirin alone, but more major bleeding recommended. At that time, the decision to continue with TAT
events occurred. The study was stopped early for superiority for up to 30 days depends on evaluation of a patient’s isch-
of the rivaroxaban plus aspirin group after a mean follow-up emic and bleeding risk. In patients with high ischemic risk

PSAP 2022 Book 1 • Cardiology 98 Antithrombotic Therapy in Cardiac Interventions


AHA/ACC Strategy for High AHA/ACC Strategy for Low
AHA/ACC Default Strategy
Ischemic/Low Bleed Risk Ischemic/High Bleed Risk

TAT up to 1 week TAT up to 1 month TAT up to 1 week


Peri-PCI to
ESC: Recommended strategy for ESC: Recommended strategy for ESC: Can discontinue aspirin
1 week
most SIHD and ACS patients most SIHD and ACS patients earlier than 1 week

1 month

3 months DAT up to 6 months


DAT up to 12 months
ESC: Consider for medically
ACC in SIHD: P2Y12 inhibitor
DAT up to 12 months managed ACS ACC in SIHD:
for 6 months, then aspirin + OAC
Discontinue P2Y12 after
6 months for 6 months
3 months

12 months

> 12 months OAC Monotherapy OAC Monotherapy OAC Monotherapy

Figure 3. Approaches to antithrombotic therapy in patients with atrial fibrillation undergoing percutaneous coronary
intervention.
ACC = American College of Cardiology; ACS = acute coronary syndrome; AHA = American Heart Association; DAT = dual
antithrombotic therapy (OAC + single antiplatelet); DAPT = dual antiplatelet therapy; ESC = European Society of Cardiology; OAC =
oral anticoagulant; PCI = percutaneous coronary intervention; SIHD = stable ischemic heart disease; TAT = triple antithrombotic
therapy (OAC + DAPT).
Information from: Angiolillo DJ, Bhatt DL, Cannon CP, et al. Antithrombotic therapy in patients with atrial fibrillation treated with oral
anticoagulation undergoing percutaneous coronary intervention: a North American perspective: 2021 update. Circulation 2021;143:
583-96; Kumbhani DJ, Cannon CP, Beavers CJ, et al. 2020 ACC expert consensus decision pathway for anticoagulant and antiplatelet
therapy in patients with atrial fibrillation or venous thromboembolism undergoing percutaneous coronary intervention or with
atherosclerotic cardiovascular disease: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll
Cardiol 2021;77:629-58; Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of atrial
fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J 2021;42:373-498.

and low bleed risk, TAT can be considered for the first month, state that ticagrelor is an acceptable alternative, but only if
followed by DAT for 6 (SIHD) to 12 (ACS) months. In patients the patient is considered a high-thrombotic, low-bleed risk.
with high bleeding risk, TAT duration can be as short as the Prasugrel should be avoided in this setting.
hospital stay, followed by DAT for 6 months. After comple-
tion of DAT, OAC monotherapy is recommended for second- Landmark Trials Assessing Dual vs. Triple
ary prevention. Table 10 summarizes current clinical practice Antithrombotic Therapy
recommendations. The concept of DAT versus TAT was first studied by the
For patients without contraindications (e.g., moderate to WOEST trial, wherein warfarin plus clopidogrel was compared
severe mitral stenosis, mechanical prosthetic valve), direct with warfarin plus DAPT. DAT was associated with less bleed-
oral anticoagulants (DOACs) are preferred for TAT and DAT. ing than TAT—without sacrificing efficacy. That paved the way
The recommendation comes from several clinical trials for further studies assessing DAT, and each of the DOACs has
demonstrating DOACs’ better safety profiles compared with had a clinical trial published in recent years. Table 11 summa-
VKA’s, including in the setting of DAT. The P2Y12 inhibitor of rizes those studies and key findings. The results of all of the
choice in this setting is clopidogrel. Compared with ticagre- studies consistently support the fact that DAT is associated
lor and prasugrel, clopidogrel has been associated with less with less bleeding—seemingly without sacrificing ischemic
bleeding (Wallentin 2009, Wiviott 2007), and an overwhelm- efficacy. In addition, the AUGUSTUS trial assessing apixaban
ing majority of patients in the clinical trials assessing TAT in DAT versus TAT established that the reduction in bleeding
received clopidogrel. Data surrounding the use of ticagrelor in fact results because of the removal of aspirin, because the
and prasugrel in this setting are limited. Current guidelines

PSAP 2022 Book 1 • Cardiology 99 Antithrombotic Therapy in Cardiac Interventions


Table 10. Summarized Clinical Practice Guideline Recommendations for Dual and Triple Antithrombotic Therapy

2021 AHA Focused 2020 ACC Expert Consensus


Update: Antithrombotic Decision Pathway for
Therapy in Patients Anticoagulant and
with AF Treated with Antiplatelet Therapy in
Oral Anticoagulation Patients with AF or VTE 2020 ESC Atrial Fibrillation
Undergoing PCI Undergoing PCI Guidelines

Oral anticoagulant DOAC DOAC DOAC


recommendation • Rivaroxaban 15 mg daily
If warfarin used, aim for If warfarin used, reasonable to and dabigatran 110 mg
a reduced target INR of target INR of 2.0–2.5 twice daily if HAS-BLED
2.0–2.5 >3 while prescribed
concomitant P2Y12 inhibitor

If VKA used, target INR of


2–2.5 (IIa-B)

Antiplatelet Aspirin + P2Y12 during peri- SIHD: SIHD:


recommendation PCI period until discharge P2Y12 inhibitor for 6 months; Aspirin + P2Y12 for 1 month;
and up to 1 week after then aspirin or clopidogrel then stop aspirin
PCI; then stop aspirin and for 6 months. Continue OAC Continue P2Y12 for 6–12
continue P2Y12 indefinitely months of total therapy
Continue OAC indefinitely
ACS:
P2Y12 inhibitor for 12 months. ACS:
Continue OAC indefinitely Aspirin + P2Y12 for 1 month;
then stop aspirin
Continue P2Y12 for 6–12
months of total therapy
Continue OAC indefinitely

Medically treated ACS:


P2Y12 for 6 months

P2Y12 of choice Clopidogrel Clopidogrel Clopidogrel


Ticagrelor acceptable in Ticagrelor acceptable in high
high ischemic risk ischemic risk

Increased thrombotic risk, Continue TAT for up to 1 Can consider TAT for up to 30 Can consider TAT for up to 1
acceptable bleeding risk month; then DAT for 6–12 days month
months; then OAC alone

Low thrombotic risk or high Discontinue P2Y12 inhibitor 6 Discontinue P2Y12 inhibitor Can consider discontinuing
bleeding risk months after PCI after 3 months for SIHD or aspirin earlier than 1 month—
after 6 months for ACS even ≤ 1 week

ACS = acute coronary syndrome, AF = atrial fibrillation; DAPT = dual antiplatelet therapy; DAT = dual antithrombotic therapy; INR =
international normalized ratio; OAC = oral anticoagulant; PCI = percutaneous coronary intervention; SIHD = stable ischemic heart dis-
ease; VKA = vitamin K antagonist; VTE = venous thromboembolism.
Information from: Angiolillo DJ, Bhatt DL, Cannon CP, et al. Antithrombotic therapy in patients with atrial fibrillation treated with oral
anticoagulation undergoing percutaneous coronary intervention: a North American perspective: 2021 update. Circulation
2021;143:583-96.
Kumbhani DJ, Cannon CP, Beavers CJ, et al. 2020 ACC expert consensus decision pathway for anticoagulant and antiplatelet
therapy in patients with atrial fibrillation or venous thromboembolism undergoing percutaneous coronary intervention or with
atherosclerotic cardiovascular disease: a report of the American College of Cardiology Solution Set Oversight Committee. J Am
Coll Cardiol 2021;77:629-58; Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of
atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J
2021;42:373-498.

PSAP 2022 Book 1 • Cardiology 100 Antithrombotic Therapy in Cardiac Interventions


Table 11. Landmark Trials Assessing Dual vs. Triple Antithrombotic Therapy

Trial (year) Antithrombotic regimens Outcomes

WOEST (2013) Warfarin + clopidogrel Compared with triple therapy, dual therapy resulted
in fewer bleeding complications—and without an
Warfarin + DAPT
increase in the rate of thrombotic events

PIONEER AF-PCI (2016) Rivaroxaban 15 mg daily (or 10 mg daily if Compared with warfarin + DAPT, rivaroxaban-based
CrCl 30–50 mL/min) + P2Y12 inhibitor antithrombotic therapies resulted in less TIMI major
and minor bleeding
Rivaroxaban 2.5 mg twice daily + DAPT for 1,
6, or 12 months
The rate of cardiovascular death, myocardial
After completion of DAPT, patients received
infarction, or stroke did not differ between the
rivaroxaban 15 mg daily + aspirin
groups
Warfarin + DAPT for 1, 6, or 12 months
After completion of DAPT, patients received
warfarin + aspirin

RE-DUAL PCI (2017) Dabigatran 110 mg twice daily + P2Y12 Compared with triple therapy using warfarin as the
inhibitor anticoagulant, dual therapy with a dabigatran-
based antithrombotic regimen resulted in reduced
Dabigatran 150 mg twice daily + P2Y12
bleeding events
inhibitor

Warfarin + P2Y12 inhibitor + aspirin Dual therapy with dabigatran 110 mg twice daily
resulted in lower rates of bleeding (15.4%) compared
with 150 mg twice daily (20.2%), though these
groups were not directly compared with one another
for noninferiority

The incidence of thromboembolic events, death, or


unplanned revascularization in the dual therapy
groups was noninferior to triple therapy

AUGUSTUS (2019) Apixaban 5 mg twice dailya + P2Y12 + aspirin Dual therapy with an apixaban-based regimen
resulted in lower bleeding compared with a
Apixaban 5 mg twice dailya + P2Y12
warfarin-based dual therapy—with lower rates of
Warfarin + P2Y12 + aspirin death or rehospitalization.
Warfarin + P2Y12
Triple therapy resulted in greater bleeding without a
difference in efficacy

ENTRUST-AF PCI (2019) Edoxaban 60 mg daily + clopidogrel Dual therapy with an edoxaban-based antithrombotic
regimen was noninferior to VKA-based triple therapy
Warfarin + clopidogrel + aspirin
with regard to clinically relevant nonmajor bleeding

The rates of cardiovascular death, myocardial


infarction, stroke, systemic embolism, or definite
stent thrombosis did not differ between the groups

a
Dose adjusted to 2.5 mg twice daily if patient met two of the following criteria, per package insert: weight <60 kg, age >80 years,
serum creatinine ≥1.5 mg/dL or greater
CrCl = creatinine clearance; DAPT = dual antiplatelet therapy; VKA = vitamin K antagonist.
Information from: Dewilde WJ, Oirbans T, Verheugt FW, et al. Use of clopidogrel with or without aspirin in patients taking oral
anticoagulant therapy and undergoing percutaneous coronary intervention: an open-label, randomised, controlled trial. Lancet
2013;381:1107-15; Gibson CM, Mehran R, Bode C, et al. Prevention of bleeding in patients with atrial fibrillation undergoing PCI. N
Engl J Med 2016;375:2423-34; Cannon CP, Bhatt DL, Oldgren J, et al. Dual antithrombotic therapy with dabigatran after PCI in atrial
fibrillation. N Engl J Med 2017;377:1513-24; Lopes RD, Heizer G, Aronson R, et al. Antithrombotic therapy after acute coronary
syndrome or PCI in atrial fibrillation. N Engl J Med 2019;380:1509-24; Vranckx P, Valgimigli M, Eckardt L, et al. Edoxaban-based
versus vitamin K antagonist–based antithrombotic regimen after successful coronary stenting in patients with atrial fibrillation
(ENTRUST-AF PCI): a randomised, open-label, phase 3b trial. Lancet 2019;394:1335-43.

PSAP 2022 Book 1 • Cardiology 101 Antithrombotic Therapy in Cardiac Interventions


combination of apixaban plus P2Y12 was associated with less
Box 2. Thrombotic and Bleeding
bleeding than was apixaban plus DAPT.
Risk Factors
Clinical Controversies with Triple Thrombosis Risk Factors
Antithrombotic Therapy
Acute coronary syndrome presentation
Recurrent MI
Even though the recommendation for DAT comes with sup- Prior stent thrombosis on antiplatelet therapy
port from strong clinical evidence, some outstanding ques- Complex PCI (three vessels treated, more than three
tions and gray areas within the application of DAT still remain. stents implanted, bifurcation with two stents implant-
ed, total stent length > 60 mm, surgical bypass graft
or CTO target lesion, atherectomy device use, LM PCI,
Ischemic vs. Bleeding Risk During the First stenting last patent vessel)
Month of Therapy Multivessel CAD
As stated previously, clinical guidelines recommend that TAT Concomitant peripheral arterial disease
be considered for up to 30 days in patients with high-throm- Premature CAD (occurring at age <45 years) or
botic-, low-bleeding-risk profiles. It is important to note that accelerated CAD (new lesion within 2 years)
Diabetes mellitus requiring therapy
all of the clinical trials assessing TAT versus DAT were pow-
CKD (eGFR <60 mL/min)
ered to detect differences in safety outcomes, such as bleed-
ing events. None of the trials were prospectively designed to Bleeding Risk Factors
Hypertension
compare ischemic efficacy, and some data suggest the pos-
Abnormal renal or liver function
sibility of increased risk of stent thrombosis and MI with DAT History of stroke or ICH
compared with TAT (Gargiulo 2019). Ischemic risk tends to be Bleeding history or bleeding diathesis (e.g., anemia with
highest in the initial period after PCI, whereas bleeding risk Hgb <11 g/dL)
is more cumulative over time (Schömig 2009). Post hoc anal- Labile INR (if on VKA)
Older >65 years old
yses from the AUGUSTUS trial demonstrated that the inci-
Drugs (concomitant OAC and antiplatelet therapy,
dence of stent thrombosis was numerically higher in patients NSAIDs)
treated with DAT and that 80% of those occurred during the Excessive alcohol consumption
first 30 days after PCI (Lopes 2020).
The authors of the AUGUSTUS trial assessed at 30 days CAD = coronary artery disease; CKD = chronic kidney disease;
CTO = chronic total occlusion; eGFR = glomerular filtration rate;
the outcomes of patients randomized to aspirin or placebo
ICH = intracranial hemorrhage; INR = international normal-
(Alexander 2020). The increase in severe bleeding with the ized ratio; LM = left main; MI = myocardial infarction; NSAID =
addition of aspirin equaled the reduction in severe ischemic nonsteroidal anti-inflammatory drug; OAC = oral anticoagula-
tion; PCI = percutaneous coronary intervention; VKA = vitamin
events during the first 30 days, but the increase in bleeding K antagonist.
persisted beyond 30 days—without any benefit in reducing Information from: Angiolillo DJ, Bhatt DL, Cannon CP, et al.
ischemic outcomes. Taking those results into consideration, Antithrombotic therapy in patients with atrial fibrillation
treated with oral anticoagulation undergoing percutaneous
recommendations for up to 30 days of TAT in patients with coronary intervention: a North American perspective: 2021
high thrombosis risk are reasonable. Risk factors for throm- update. Circulation 2021;143:583-96; Hindricks G, Potpara
bosis and bleeding are given in Box 2. Overall, an individu- T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and
management of atrial fibrillation developed in collaboration
alized approach to antithrombotic therapy during the first with the European Association for Cardio-Thoracic Surgery
month after PCI is essential. Beyond 30 days, DAT is likely the (EACTS). Eur Heart J 2021;42:373-498.
appropriate treatment strategy for most patients.

Novel P2Y12 Inhibitor Use


Guidelines recommend that ticagrelor be used in patients
ischemic and low bleed risk, but more data are needed spe-
with high-ischemic-, low-bleeding-risk profiles. It is import-
cific to patients requiring OAC.
ant to note that the data assessing this particular P2Y12
The data for prasugrel use in patients receiving OAC are
inhibitor in the setting of TAT are limited. In landmark trials
sparse. When used in the setting of TAT, prasugrel has been
(see Table 11), ticagrelor was used in up to 12% of patients.
associated with a fourfold increase in bleeding (Sarafoff
Ticagrelor has demonstrated superiority to clopidogrel in
2013). As a result, guidelines recommend avoiding prasugrel,
reducing ischemic events in ACS, but its increased potency
but more data are needed to evaluate prasugrel’s role in the
raises concerns about increased bleeding (Wallentin 2009).
setting of DAT.
Indeed, in the setting of concomitant OAC use, ticagrelor has
demonstrated increased bleeding compared with clopido-
Secondary Prevention in Patients Requiring
grel (Andreou 2018). Based on the existing evidence, it is rea-
Anticoagulation
sonable to consider ticagrelor in treating patients with high
Clinical Guideline Recommendations

PSAP 2022 Book 1 • Cardiology 102 Antithrombotic Therapy in Cardiac Interventions


Table 12. Landmark Trials Assessing OAC Monotherapy for Secondary Prevention

Trial Antithrombotic Regimens Outcomes

AFIRE (2019) a
Rivaroxaban 15 mg daily (10 mg daily for CrCl Trial terminated early based on excessive
15–49 mL/min) mortality in rivaroxaban + antiplatelet arm
Vs. (1.9% vs. 3.4%, p<0.05)
Rivaroxaban 15 mg (10 mg daily for CrCl
15–49 mL/min) + antiplatelet Rivaroxaban alone was noninferior for ischemic
outcomes and associated with less major
bleeding

OAC-ALONE (2019)a OAC monotherapy Trial terminated early based on slow enrollment
Vs.
OAC + antiplatelet Noninferiority of OAC alone was not established
for primary ischemic end point—likely because
of inadequate power

Significantly higher rates of major bleeding were


noted with OAC + antiplatelet

a
Both studies were conducted in Japan, so dosing strategies differ from US FDA–approved dosing.
CrCl = creatinine clearance; OAC = oral anticoagulation.
Information from: Matsumura-Nakano Y, Shizuta S, Komasa A, et al. Open-label randomized trial comparing oral anticoagulation
with and without single antiplatelet therapy in patients with atrial fibrillation and stable coronary artery disease beyond 1 year after
coronary stent implantation. Circulation 2019;139:604-16; Yasuda S, Kaikita K, Akao M, et al. Antithrombotic therapy for atrial
fibrillation with stable coronary disease. N Engl J Med 2019;381:1103-13.

The most recent guidelines from the AHA, ACC, and ESC all overwhelming majority of patients develop AS secondary to
recommend that patients with stable coronary artery disease degeneration of the aortic valve, but rheumatic heart disease
requiring no further intervention beyond 12 months receive and congenital bicuspid aortic valve are other etiologies of
oral anticoagulation monotherapy for secondary disease pre- AS, particularly in younger patients. Clinical risk factors for
vention (Angiolillo 2021; Hindricks 2021; Kumbhani 2021). AS mirror those of CAD, including older age, male sex, ele-
The statements cite the AFIRE and OAC-ALONE trials as evi- vated LDL, hypertension, smoking, diabetes, and metabolic
dence to support OAC monotherapy (Table 12) (Matsumura- syndrome. Patients with histories of mediastinal irradiation,
Nakano 2019, Yasuda 2019). However, in patients with high renal failure, familial hypercholesterolemia, or disorders of
thrombotic risk and low bleeding risk (see Table 12), it may be calcium metabolism are also at increased risk of developing
AS.
reasonable to consider aspirin in addition to OAC for second-
Aortic stenosis is associated with several cardiac struc-
ary prevention (Kumbhani 2021).
tural changes, including left ventricular hypertrophy, dia-
stolic dysfunction, and decreased longitudinal shortening,
ANTITHROMBOTIC THERAPY IN although the ejection fraction typically remains normal
TRANSCATHETER AORTIC VALVE (Otto 2014). If left untreated, the resulting obstruction of
IMPLANTATION left ventricular outflow leads to inadequate cardiac output,
decreased exercise capacity, heart failure, and death from
Overview of Aortic Stenosis
cardiovascular causes. The classic triad of AS symptoms
Aortic stenosis (AS) is a progressive, chronic valvular dis-
consist of angina, dyspnea, and syncope and are typically
ease characterized by a narrowing of the aortic valve open- not present until late in the disease. Once a patient devel-
ing (Otto 2014). The spectrum of disease spans from mild ops severe, symptomatic AS, mortality is greater than 50%
fibrocalcific changes in the leaflets to obstruction of left ven- at 2 years if the valve is not replaced. Aortic valve replace-
tricular outflow. Endothelial disruption with inflammation ment remains the gold standard of treatment because no
and lipid infiltration initiates this disease, ultimately lead- medical therapies have been proven to slow the progres-
ing to tissue calcification and valve obstruction. The preva- sion of or reverse AS. Current guidelines make a Class I rec-
lence of AS increases with age, occurring in 0.2% of adults ommendation for aortic valve replacement in patients with
50–59 years and in 9.8% of adults 80–89 years. An severe, symptomatic AS and in those without symptoms but

PSAP 2022 Book 1 • Cardiology 103 Antithrombotic Therapy in Cardiac Interventions


with ejection fractions of less than 50% or who are under- Clinical Guideline Recommendations
going coronary artery bypass graft (Otto 2021). For several for Antithrombotic Therapy After TAVI
years, surgical aortic valve replacement (SAVR) was the only Antithrombotic therapy after TAVI has been an evolving
approach to aortic valve replacement. clinical practice since its implementation. Initially, studies
designed to evaluate TAVI technology defaulted to DAPT. Rea-
Transcatheter-Aortic-Valve-Implantation sons that DAPT was the initial chosen antithrombotic strat-
Indication and Procedure egy include the assumption of blood flow conditions similar
Transcatheter aortic valve implantation (TAVI)—also known to those with coronary stents, similar to the goal of prevent-
as transcatheter aortic valve replacement, or TAVR—recently ing device-related thromboembolic events, and similar to the
emerged as a less-invasive alternative to SAVR for the treat- need to protect the metallic valve frame during endotheliali-
ment of AS. Transcatheter aortic valve implantation is rec- zation (Guedeney 2019). However, recent literature has chal-
ommended in patients older than 65 years who have severe, lenged that empiric recommendation because it has not been
symptomatic AS, and shared decision making should deter- studied robustly and does not apply to patients requiring
mine whether TAVI or SAVR is the more appropriate AVR chronic OAC.
approach (Otto 2021). It is important to note that for patients The 2020 AHA/ACC Valvular Heart Disease Guidelines
with severe AS and prohibitive surgical risk, TAVI offers an recommend low-dose aspirin 75–100 mg daily for patients
option for valve replacement because it has demonstrated undergoing TAVI in the absence of other indications for OAC
improved mortality compared with management by means (Class 2A, LOE B-R) (Otto 2021). They also suggest the addi-
of balloon valvuloplasty or medical therapy (Leon 2010). The tion of clopidogrel for 3–6 months in patients at low risk of
TAVI procedure involves percutaneous insertion of a bio- bleeding (Class 2B, LOE B-NR). It is important to note that
prosthetic valve through either balloon expansion or a self- these guidelines do not specifically address antithrombotic
expanding valve—without removing the calcified native valve. therapy in patients requiring chronic OAC, nor do they include
Published literature surrounding TAVI involves the transfemo- data from the POPular-TAVI trials—the largest randomized tri-
ral approach, though transapical and transaortic approaches als assessing single antiplatelet therapy (SAPT) versus DAPT
have been described but may carry a higher risk of mortality. and OAC with or without antiplatelet therapy. The ESC 2021

OAC Indication No OAC Indication


Pre-TAVI

OAC Single Antiplatelet

Unfractionated Unfractionated
heparin heparin

Goal ACT ≥250–300s Goal ACT ≥250–300s


TAVI

Coronary stent <3 Coronary stent <3


months ago months ago

Yes No Yes No
Post-TAVI

Dual Therapy
1– 6 Months

Aspirin + Clopidogrel
OAC + Clopidogrel or
Aspirin
VKA or NOAC Aspirin OR
Monotherapy Clopidogrel
Lifelong

VKA or NOAC Aspirin OR


Monotherapy Clopidogrel

Figure 4. European Society of Cardiology Recommendations for Antithrombotic treatment before and after TAVI.
ACT = activated clotting time; NOAC = novel oral anticoagulant; OAC = oral anticoagulant; TAVI = transaortic valvular intervention.
Information from: Ten Berg J, Sibbing D, Rocca B, et al. Management of antithrombotic therapy in patients undergoing transcatheter
aortic valve implantation: a consensus document of the ESC Working Group on Thrombosis and the European Association of
Percutaneous Cardiovascular Interventions (EAPCI), in collaboration with the ESC Council on Valvular Heart Disease. Eur Heart J
2021;42:2265-9.

PSAP 2022 Book 1 • Cardiology 104 Antithrombotic Therapy in Cardiac Interventions


Consensus Statement on antithrombotic therapy after TAVI The results of the aforementioned studies are cited by
is presented in Figure 4 and presents more-thorough recom- the ESC to support the recommendation of SAPT mono-
mendations depending on whether or not a patient requires therapy after TAVI in patients without indications for OAC
OAC (Ten Berg 2021). They favor SAPT and OAC monotherapy who have not undergone recent coronary stenting (Ten
in the absence of recent coronary stenting. With the results of Berg 2021). In patients with recent coronary stents before
the POPular-TAVI trials and the publication of the recent 2021 or after TAVI, DAPT is the preferred treatment strategy for
ESC consensus statement, it is likely that clinical practice in durations described previously in this chapter. Anticoagu-
the United States will shift towards utilizing SAPT and OAC lation should not be used after TAVI unless there is another
monotherapy for most patients after TAVI. indication, such as atrial fibrillation or recent venous
thromboembolism.
Antithrombotic Therapy Without an Indication
Antithrombotic Therapy with an Indication
for OAC: Literature Review
for OAC: Literature Review
The first clinical trial designed to prospectively evaluate SAPT
Antithrombotic therapy in patients with indications for OAC
versus DAPT after TAVI was the ARTE Trial (Rodés-Cabau
(i.e., atrial fibrillation, recent venous thromboembolism) has
2017). A total of 222 patients were randomized to receive
been prospectively evaluated in only one study: the POPu-
either low-dose aspirin alone or aspirin plus clopidogrel after
lar TAVI Cohort B (Nijenhuis 2020). In this arm of the POPu-
TAVI. The composite end point of death, MI, stroke, or tran-
lar TAVI trial, 326 patients requiring OAC were randomized to
sient ischemic attack or a major or life-threatening bleeding
receive either clopidogrel or placebo for 3 months in addition
tended to occur more often in the DAPT group (15.3% vs. 7.2%;
to OAC. The majority of patients (95%) in this study had atrial
p=0.065); and the lack of statistical significance may have
fibrillation as their indication for OAC. All bleeding and non-
been the result of the small sample size and early termination
procedural-related bleeding events were significantly reduced
because of slow enrollment. It is important that the rates of
in the OAC group compared with OAC plus clopidogrel (all
death, MI, stroke, and transient ischemic attack appeared to
bleeding: 21.7% vs. 34.6%; p=0.01; non-procedural-related
be similar between the groups, whereas major and life-threat-
bleeding: 21.7% vs. 34.0%; p=0.02), and most of the bleeding
ening bleeding occurred more often in the DAPT group (10.8%
events were minor and occurred during the first month of ther-
vs. 3.6%; p=0.038).
apy. Ischemic outcomes were similar between the two groups.
The superior safety of SAPT compared with DAPT after
The ESC guidelines do not give preference to DOAC or VKA
TAVI was confirmed by the POPular TAVI Cohort A (POPular-
after TAVI, and there are only limited comparative data in this
TAVI-A) trial in 2020 (Brouwer 2020), a study designed sim-
setting (Ten Berg 2021). In POPular TAVI-B, nearly 70% of the
ilarly to the ARTE trial: patients were randomized to receive
patients were receiving a VKA as their anticoagulant. Obser-
either low-dose aspirin or aspirin plus clopidogrel for 3 months
vational data show inconsistent results for DOACs in AF after
after TAVI. However, POPular-TAVI-A comprised nearly three
TAVI. A German registry of more than 900 patients demon-
times the number of patients as the ARTE trial did, with 331
strated higher ischemic events at 1 year with DOAC use, and
patients randomized to receive SAPT and 334 randomized to
a Danish registry showed similar rates between DOACs and
receive DAPT. All of the primary outcomes in the POPular-TA-
VKA (Butt 2021; Jochheim 2019). In clinical practice, DOACs
VI-A trial were bleedings (minor, major, and life-threatening)
continue to be used very often in patients with AF undergoing
and non-procedure-related bleedings at 12 months, both of
TAVI, and the risk of ischemic events in DOACs versus VKA
which occurred significantly less in the SAPT arm compared
warrants further study.
with DAPT. Ischemic outcomes—including death from cardio-
As a result of the randomized POPular-TAVI-B data, the
vascular causes, MI, and stroke—were similar between the
ESC recommends OAC monotherapy in patients with indica-
two groups.
tions for OAC who are undergoing TAVI (Ten Berg 2021). In
After TAVI, anticoagulation in patients without any other
patients who have also undergone recent coronary stenting,
indications for anticoagulation was evaluated in the GALILEO
a dual-therapy approach that uses OAC plus an antiplatelet
study (Dangas 2020) and the ATLANTIS trial (pending pub-
as described previously is recommended, followed by OAC
lication). GALILEO terminated early because safety was of
monotherapy for secondary prevention.
concern based on the fact that the rivaroxaban arm was asso-
ciated with a higher risk of death or thromboembolic compli-
ANTITHROMBOTIC THERAPY
cations as well as a higher risk of bleeding compared with
IN MITRACLIP
DAPT. As a result, both the AHA/ACC and ESC guidelines
strongly recommend against the use of rivaroxaban after Mitral Regurgitation Overview

TAVI in the absence of other indications for anticoagulation. Mitral regurgitation (MR) is a result of retrograde blood flow
ATLANTIS failed to show any benefit with apixaban compared from the left ventricle into the left atrium through the mitral
with antiplatelet therapy. valve. With a prevalence that increases with age, MR is the
most common type of moderate or severe valvular heart

PSAP 2022 Book 1 • Cardiology 105 Antithrombotic Therapy in Cardiac Interventions


Patient Care Scenario
A 76-year-old woman presents to the ED after a witnessed clopidogrel 75 mg daily, metoprolol 25 mg twice daily, and
syncopal event. She reports walking around her house losartan 25 mg daily. A transthoracic echocardiogram is
trying to clean up before her daughter came over, when performed and identifies severe, critical aortic stenosis.
her husband witnessed her fall. She reports briefly los- Because of the patient’s high surgical risk, the patient
ing consciousness but came to shortly thereafter. She is scheduled for transcatheter aortic valve implantation
did not hit her head, and CT head was negative for any for the next day. She undergoes successful TAVI and is
intracranial bleed. Her medical history is significant for admitted to the cardiology step-down floor for obser-
atrial fibrillation, CAD status post-PCI with drug-eluting vation postprocedure. What are the most appropriate
stent about 2 months prior for ongoing angina, hyper- antithrombotic regimen and duration of therapy for this
tension, and chronic kidney disease. Home medications patient after her TAVI?
are apixaban 5 mg twice daily, atorvastatin 80 mg daily,
ANSWER
This patient undergoing TAVI has two factors to consider: of home apixaban and clopidogrel—is the most appropri-
the need for oral anticoagulation for atrial fibrillation and ate antithrombotic regimen for this patient after her TAVI.
the need for DAPT after coronary stenting. According to With regard to duration of therapy, because placement of
statements from the American Heart Association, the the drug-eluting stent was in the setting of stable isch-
American College of Cardiology, and the European Society emic heart disease, the recommended duration of therapy
of Cardiology, triple antithrombotic therapy with antico- is 6 months. After completion, it is recommended that the
agulation plus DAPT should be avoided. A combination patient continue with OAC monotherapy for secondary
of OAC and a P2Y12 inhibitor—in this case continuation prevention.

1. Angiolillo DJ, Bhatt DL, Cannon CP, et al. Antithrombotic therapy in patients with atrial fibrillation treated with oral anticoagulation
undergoing percutaneous coronary intervention: a North American perspective: 2021 update. Circulation 2021;143:583-96.
2. Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of atrial fibrillation developed in collab-
oration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J 2021;42:373-498.
3. Kumbhani DJ, Cannon CP, Beavers CJ, et al. 2020 ACC expert consensus decision pathway for anticoagulant and antiplatelet therapy
in patients with atrial fibrillation or venous thromboembolism undergoing percutaneous coronary intervention or with atheroscle-
rotic cardiovascular disease: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol
2021;77:629-58.
4. Levine GN, Bates ER, Bittl JA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with
coronary artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice
Guidelines. J Am Coll Cardiol 2016;68:1082-115.
5. Ten Berg J, Sibbing D, Rocca B, et al. Management of antithrombotic therapy in patients undergoing transcatheter aortic valve implan-
tation: a consensus document of the ESC Working Group on Thrombosis and the European Association of Percutaneous Cardiovascular
Interventions (EAPCI), in collaboration with the ESC Council on Valvular Heart Disease. Eur Heart J 2021;42:2265-9.

disease among US adults older than 55 years (Nkomo 2006). left ventricle leads to dilatation and decreased contractil-
Chronic MR may present as one of two types—either chronic ity, resulting in a reduction of ejection fraction (Apostolidou
primary MR or chronic secondary MR—with each having fea- 2017).
tures related to valvular anatomy and hemodynamics, which
facilitate appropriate diagnostic staging and determination MitraClip Indication and Procedure
of treatment options. Primary MR, also called degenerative Transcatheter mitral valve repair (TMVr) with the MitraClip
MR or organic MR, results from structural deformity or dam- device is indicated for the treatment of both chronic pri-
age to the leaflets, chordae, and/or papillary muscles that mary MR and chronic secondary MR and is currently the only
causes the leaflets to close insufficiently during systole. Sec- US FDA–approved device for TMVr. The MitraClip device is
ondary MR, also called ischemic MR or functional MR, is not inserted percutaneously and is less invasive compared with
the result of a structural abnormality of the valve itself but conventional cardiac surgery for mitral valve repair. Access
is, rather, a result of left-ventricular-wall-motion abnormali- to the mitral valve is gained through the femoral vein, where a
ties or left-ventricular remodeling resulting in improper clo- catheter is threaded into the left atrium by way of transseptal
sure of the valve during the cardiac cycle (Nishimura 2014). access with the assistance of fluoroscopy and transesopha-
Over time, continued retrograde blood flow leads to volume geal echocardiography. The MitraClip is inserted through the
overload and causes ventricular dilatation, widening of the catheter and deployed upon access to the mitral valve, pulling
mitral annulus, and diminished coaptation of leaflets, lead- the front and back leaflets of the mitral valve, clipping them
ing to further worsening of MR. Eventually, volume overload together, and thus enabling the valve to close appropriately
becomes so severe that wall-stress-related afterload on the during the cardiac cycle (Feldman 2011).

PSAP 2022 Book 1 • Cardiology 106 Antithrombotic Therapy in Cardiac Interventions


Transcatheter mitral valve repair has also been associated most-recent clinical trials (Feldman 2005). However, further
with improved outcomes compared with surgical repair— studies are required to assess the use of other antiplatelet
specifically, reductions in death and surgery for valve dys- agents such as ticagrelor and prasugrel in this setting.
function (Feldman 2011)—and is a reasonable alternative to
surgical repair. It is currently recommended in patients with
CONCLUSION
chronic, severe secondary MR related to left ventricular sys-
tolic dysfunction who have persistent symptoms despite The pathophysiology by which thrombotic events occur
guideline-directed medical therapy (GDMT) (Otto 2020). Com- during and after cardiac interventions is complex, and the
pared with GDMT alone, TMVr with Mitraclip offers improve- optimization of antithrombotic therapy pre- and postcar-
ment in survival, hospitalizations, symptoms, and quality of diac intervention is an essential aspect of care. It is imper-
life (Stone 2018). ative that thrombotic and bleeding risks be evaluated so as
to be able to select the best therapy and duration of chronic
treatment. Pharmacists can play a critical role in assisting
Antithrombotic Therapy After MitraClip
Among studies assessing the MitraClip device in TMVr, the
incidence of stroke or transient ischemic attack occurred in
up to 4.4% of patients (Nusca 2018; Stone 2018). However, Practice Points
no evidence-based guidelines exist to guide choice or dura-
• Coronary stents have evolved significantly since their intro-
tion of antithrombotic regimens. Similarly, standard anti- duction in the mid-1980s, and new features in their design,
thrombotic therapy neither before nor during the procedure structure, and material continue to expand.
has been established; nor has the role of antiplatelet agent • Stent thrombosis and in-stent restenosis continue to be
loading doses before device implantation. Patients who are significant complications after coronary stent placement.
DAPT has helped significantly reduce the incidence of ST,
chronically treated with antiplatelet therapy typically con-
and the development of DES has reduced the incidence of
tinue without interruption periprocedurally (Guyatt 2012).
ISR.
Patients on oral anticoagulation should be managed the • All patients who undergo PCI should be treated with DAPT
same as for other percutaneous interventions, which is, typ- unless also receiving oral anticoagulant treatment. The
ically, a short period of anticoagulation interruption to pre- exact agent and duration of therapy depend on various
vent bleeding. patient-specific factors.
• When it comes to a switch between different P2Y12
The manufacturer of MitraClip does not provide recom-
inhibitors, several factors must be considered, including
mendations on the choice or duration of antithrombotic drug-drug interactions, timing of index event, and reloading
therapy postprocedure. The series of EVEREST studies main- of oral P2Y12 inhibitors.
tained a study protocol that consisted of a regimen of aspi- • TAT (OAC plus DAPT) can be considered for up to 30 days
rin 325 mg daily for 6–12 months in addition to clopidogrel after PCI, but a dual-therapy (OAC-plus-P2Y12 inhibitor)
75 mg daily for 1 month (Whitlow 2012; Feldman 2011; Mauri approach is generally safer for most patients. Ultimately,
the approach to antithrombotic therapy in patients
2010; Feldman 2005). The ACCESS-EU study used a regimen
requiring both OAC and DAPT should be individualized
of aspirin 100 mg daily for 3 months in addition to clopidogrel based on an individual patient’s ischemic and bleeding risk.
75 mg daily for 4 weeks, and the COAPT trial used a clopido- • Clopidogrel remains the P2Y12 inhibitor of choice in the
grel loading dose (≥300 mg) within 24 hours before procedure setting of TAT, but ticagrelor can be considered in patients
(6–24 hours if possible) or immediately after the procedure with high ischemic risks who present with an acute
coronary syndrome. Prasugrel should generally be avoided
(Maisano 2013). Aspirin was used at the proceduralist’s dis-
in patients requiring anticoagulation.
cretion, with a recommended loading dose of aspirin 325 mg • The evidence surrounding antithrombotic therapy after
before or immediately after the procedure—if used. Postproce- TAVI has grown in the past few years and favors SAPT
dural antithrombotic therapy consisted of clopidogrel 75 mg or OAC monotherapy, as stated in the ESC consensus
daily with or without aspirin 81 mg daily for 6 months or lon- statement.
ger. If patients were previously taking chronic oral anticoag- • Because of increased harm in this setting, anticoagulation
should not be used after TAVI unless there is another
ulation (warfarin or DOAC), the anticoagulant was continued
indication, such as atrial fibrillation or venous
without antiplatelet therapy (Stone 2018). thromboembolism.
The 2020 ACC/AHA Guideline for the Management of • No evidence-based guidelines have been currently
Patients with Valvular Heart Disease provides no input with published for guidance after MitraClip implantation, and
regard to antithrombotic management surrounding Mitra- the existing literature specifies a combination of aspirin
and clopidogrel. Exact dosing and duration may be left to
Clip placement, nor do the 2017 ESC Valvular Heart Disease
the discretion of the interventionalists.
Guidelines. Therefore, it is reasonable that antiplatelet-na-
• Clinical pharmacists have a vital role to play in ensur-
ive patients may initiate aspirin and clopidogrel immedi- ing optimal antithrombotic therapy after PCI, TAVI, and
ately after the procedure given the study protocols in the Mitraclip.

PSAP 2022 Book 1 • Cardiology 107 Antithrombotic Therapy in Cardiac Interventions


with the selection of, duration of, and transition between with non-ST elevation acute coronary syndrome. Kardiol
P2Y12 inhibitors for coronary stenting in both SIHD and Pol 2015;73:592-7.
ACS. For patients requiring both anticoagulation and treat- Brouwer J, Nijenhuis VJ, Delewi R, et al. Aspirin with or with-
ment with antiplatelet therapy, pharmacists can apply the out clopidogrel after transcatheter aortic-valve implanta-
approach of DAT—anticoagulation plus P2Y12 inhibitor—to tion. N Engl J Med 2020;383:1447-57.
reduce bleeding risk while maintaining ischemic efficacy.
Butt JH, De Backer O, Olesen JB, et al. Vitamin K antagonists
The approach to antithrombotic therapy in TAVI continues vs. direct oral anticoagulants after transcatheter aortic
to evolve, but current literature and consensus statements valve implantation in atrial fibrillation. Eur Heart J Cardio-
suggest that SAPT and OAC monotherapy are appropriate for vasc Pharmacother 2021;7:11-9.
most patients. Last, with regard to MitraClip, further data are
Claassens DM, Sibbing D. De-escalation of antiplatelet treat-
needed to better define the optimal agents and duration, but
ment in patients with myocardial infarction who under-
DAPT with aspirin and clopidogrel is reasonable. Ultimately, went percutaneous coronary intervention: a review of the
pharmacists maintain an essential role in the selection of current literature. J Clin Med 2020;9:2983.
antithrombotic therapies to mitigate bleeding and prevent
Collet J-P, Thiele H, Barbato E, et al. 2020 ESC guidelines for
ischemic-related events.
the management of acute coronary syndromes in patients
presenting without persistent ST-segment elevation.
REFERENCES Eur Heart J 2021;42:1289-367.
ACTIVE Writing Group of the ACTIVE Investigators, Connolly
Dangas GD, Claessen BE, Caixeta A, et al. In-stent reste-
S, Pogue J, et al. Clopidogrel plus aspirin versus oral anti-
nosis in the drug-eluting stent era. J Am Coll Cardiol
coagulation for atrial fibrillation in the atrial fibrillation
2010;56:1897-907.
clopidogrel trial with irbesartan for prevention of vascu-
lar events (active w): a randomised controlled trial. Lancet Dangas GD, Tijssen JGP, Wöhrle J, et al. A controlled trial of
2006;367:1903-12. rivaroxaban after transcatheter aortic-valve replacement.
N Engl J Med 2020;382:120-9.
Adriaenssens T, Byrne R. Prevention of late stent thrombosis
by an interdisciplinary global European effort: PRESTIGE. Dewilde WJ, Oirbans T, Verheugt FW, et al. Use of clopido-
Eur Heart J 2014;35:2128-9. grel with or without aspirin in patients taking oral antico-
agulant therapy and undergoing percutaneous coronary
Alexander JH, Wojdyla D, Vora AN, et al. Risk/benefit tradeoff
intervention: an open-label, randomised, controlled trial.
of antithrombotic therapy in patients with atrial fibrillation
Lancet 2013;381:1107-15
early and late after an acute coronary syndrome or percu-
taneous coronary intervention: insights from AUGUSTUS. Eikelboom JW, Connolly SJ, Bosch J, et al. Rivaroxaban with
Circulation 2020;141:1618-27. or without aspirin in stable cardiovascular disease. N Engl
J Med 2017;377:1319-30.
Amsterdam EA, Wenger NK, Brindis RG, et al. 2014 ACC/
AHA guideline for the management of patients with non– Erlinge D, Ten Berg J, Foley D, et al. Reduction in platelet
ST-elevation acute coronary syndromes: a report of the reactivity with prasugrel 5 mg in low-body-weight patients
American College of Cardiology/American Heart Asso- is noninferior to prasugrel 10 mg in higher-body-weight
ciation Task Force on Practice Guidelines. Circulation patients: results from the FEATHER trial. J Am Coll Cardiol
2014;130:e344-e426. 2012;60:2032-40.
Andreou I, Briasoulis A, Pappas C, et al. Ticagrelor versus Feldman T, Foster E, Glower DD, et al. Percutaneous
clopidogrel as part of dual or triple antithrombotic ther- repair or surgery for mitral regurgitation. N Engl J Med
apy: a systematic review and meta-analysis. Cardiovasc 2011;364:1395-1406.
Drugs Ther 2018;32:287-94.
Feldman T, Wasserman HS, Herrmann HC, et al. Percutane-
Angiolillo DJ, Bhatt DL, Cannon CP, et al. Antithrombotic ous mitral valve repair using the edge-to-edge technique:
therapy in patients with atrial fibrillation treated with oral six-month results of the EVEREST phase I clinical trial.
anticoagulation undergoing percutaneous coronary inter- Am Coll Cardiol 2005;46:2134-40.
vention: a North American perspective: 2021 update.
Circulation 2021;143:583-96. Fihn SD, Gardin JM, Abrams J, et al. 2012 ACCF/AHA/ACP/
AATS/PCNA/SCAI/STS guideline for the diagnosis and
Angiolillo DJ, Rollini F, Storey RF, et al. International expert management of patients with stable ischemic heart
consensus on switching platelet P2Y12receptor–inhibiting disease. J Am Coll Cardiol 2012;60:e44-164.
therapies. Circulation 2017;136:1955-75.
Franchi F, Rollini F, Rivas Rios J, et al. Pharmacodynamic
Apostolidou E, Maslow AD, Poppas A. Primary mitral valve effects of switching from ticagrelor to clopidogrel in
regurgitation: update and review. Glob Cardiol Sci Pract patients with coronary artery disease: results of the
2017;2017:e201703. swap-4 study. Circulation 2018;137:2450-62.
Avci BK, Ikitimur B, Tok OO, et al. The role of GRACE score in Furtado RHM, Nicolau JC, Guo J, et al. Morphine and cardio-
the prediction of high-risk coronary anatomy in patients vascular outcomes among patients with non-ST-segment

PSAP 2022 Book 1 • Cardiology 108 Antithrombotic Therapy in Cardiac Interventions


elevation acute coronary syndromes undergoing coronary Kubica J, Adamski P, Ostrowska M, et al. Morphine delays
angiography. J Am Coll Cardiol 2020;75:289-300. and attenuates ticagrelor exposure and action in patients
with myocardial infarction: the randomized, double-blind,
Gargiulo G, Goette A, Tijssen J, et al. Safety and efficacy placebo-controlled IMPRESSION trial. Eur Heart J
outcomes of double vs. triple antithrombotic therapy 2016;37:245-52.
in patients with atrial fibrillation following percutaneous
coronary intervention: a systematic review and Kumbhani DJ, Cannon CP, Beavers CJ, et al. 2020 ACC
meta-analysis of non–vitamin K antagonist oral expert consensus decision pathway for anticoagulant
anticoagulant-based randomized clinical trials. Eur Heart and antiplatelet therapy in patients with atrial fibrillation
J 2019;40:3757-67. or venous thromboembolism undergoing percutaneous
coronary intervention or with atherosclerotic cardiovas-
Guedeney P, Mehran R, Collet J-P, et al. Antithrombotic ther- cular disease: a report of the American College of Cardiol-
apy after transcatheter aortic valve replacement. Circ Car- ogy Solution Set Oversight Committee. J Am Coll Cardiol
diovasc Interv 2019;12: e007411. 2021;77:629-58.
Guerra E, Ndrepepa G, Schulz S, et al. Impact of inhospital Leon MB, Smith CR, Mack M, et al. Transcatheter aortic-valve
stent thrombosis and cerebrovascular accidents on long- implantation for aortic stenosis in patients who cannot
term prognosis after percutaneous coronary intervention. undergo surgery. N Engl J Med 2010;363:1597-1607.
Am Heart J 2014;168:862-8.e1.
Levine GN, Bates ER, Bittl JA, et al. 2016 ACC/AHA guideline
Guyatt GH, Akl EA, Crowther M, et al. Introduction to the focused update on duration of dual antiplatelet therapy
ninth edition: Antithrombotic Therapy and Prevention of in patients with coronary artery disease: a report of the
Thrombosis, 9th ed: American College of Chest Physi- American College of Cardiology/American Heart Associa-
cians evidence-based clinical practice guidelines. Chest tion Task Force on Clinical Practice Guidelines. J Am Coll
2012;141(2 Suppl):48S-52S. Cardiol 2016;68:1082-115.
Hansen ML, Sørensen R, Clausen MT, et al. Risk of bleed- Lopes RD, Leonardi S, Wojdyla DM, et al. Stent thrombosis in
ing with single, dual, or triple therapy with warfarin, aspi- patients with atrial fibrillation undergoing coronary stent-
rin, and clopidogrel in patients with atrial fibrillation. Arch ing in the AUGUSTUS trial. Circulation 2020;141:781-3.
Intern Med 2010;170:1433-41.
Maisano F, Franzen O, Baldus S, et al. Percutaneous mitral
Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines valve interventions in the real world. J Am Coll Cardiol
for the diagnosis and management of atrial fibrillation 2013;62:1052-61.
developed in collaboration with the European Associa-
tion for Cardio-Thoracic Surgery (EACTS). Eur Heart J Matsumura-Nakano Y, Shizuta S, Komasa A, et al. Open-label
2021;42:373-498. randomized trial comparing oral anticoagulation with and
without single antiplatelet therapy in patients with atrial
Howard CE, Nambi V, Jneid H, et al. Extended duration of fibrillation and stable coronary artery disease beyond
dual-antiplatelet therapy after percutaneous coronary 1 year after coronary stent implantation. Circulation
intervention: how long is too long? J Am Heart Assoc 2019;139:604-16.
2019;8:e012639.
Mauri L, Garg P, Massaro JM, et al. The EVEREST II trial:
Iqbal J, Gunn J, Serruys PW. Coronary stents: historical design and rationale for a randomized study of the
development, current status and future directions. Br Med evalve MitraClip system compared with mitral valve sur-
Bull 2013;106:193-211. gery for mitral regurgitation. American Heart Journal
January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS 2010;160:23-9.
focused update of the 2014 AHA/ACC/HRS guideline for McEvoy JW, Ibrahim K, Kickler TS, et al. Effect of intrave-
the management of patients with atrial fibrillation: a report nous fentanyl on ticagrelor absorption and platelet inhibi-
of the american college of cardiology/american heart tion among patients undergoing percutaneous coronary
association task force on clinical practice guidelines and intervention: the PACIFY randomized clinical trial (platelet
the heart rhythm society in collaboration with the society aggregation with ticagrelor inhibition and fentanyl). Circu-
of thoracic surgeons. Circulation 2019;140:e125-51. lation 2018;137:307-9.
Jochheim D, Barbanti M, Capretti G, et al. Oral anticoagu- Montalescot G, Bolognese L, Dudek D, et al. Pretreatment
lant type and outcomes after transcatheter aortic valve with prasugrel in non–ST-segment elevation acute coro-
replacement. JACC Cardiovasc Interv 2019;12:1566-76. nary syndromes. N Engl J Med 2013;369:999-1010.
Knuuti J, Wijns W, Saraste A, et al. 2019 ESC guidelines for Neumann FJ, Gawaz M, Ott I, et al. Prospective evaluation
the diagnosis and management of chronic coronary syn- of hemostatic predictors of subacute stent thrombosis
dromes. Eur Heart J 2020;41:407-77. after coronary Palmaz-Schatz stenting. J Am Coll Cardiol
Kogame N, Guimarães PO, Modolo R, et al. Aspirin-free pra- 1996;27:15-21.
sugrel monotherapy following coronary artery stenting in Neumann F-J, Sousa-Uva M, Ahlsson A, et al. 2018 ESC/
patients with stable cad. JACC: Cardiovascular Interven- EACTS guidelines on myocardial revascularization. Eur
tions 2020;13:2251-62. Heart J 2019;40:87-165.

PSAP 2022 Book 1 • Cardiology 109 Antithrombotic Therapy in Cardiac Interventions


Nijenhuis VJ, Brouwer J, Delewi R, et al. Anticoagulation with Sibbing D, Aradi D, Jacobshagen C, et al. Guided de-escala-
or without clopidogrel after transcatheter aortic-valve tion of antiplatelet treatment in patients with acute cor-
implantation. N Engl J Med 2020;382:1696-1707. onary syndrome undergoing percutaneous coronary
intervention (TROPICAL-ACS): a randomised, open-label,
Nishimura RA, Otto CM, Bonow RO, et al. American College multicentre trial. Lancet 2017;390:1747-57.
of Cardiology/American Heart Association Task Force on
Practice Guidelines. 2014 AHA/ACC guideline for the man- Stefanini GG, Holmes DR. Drug-eluting coronary-artery
agement of patients with valvular heart disease: executive stents. N Engl J Med 2013;368:254-65.
summary: a report of the American College of Cardiology/
American Heart Association Task Force on Practice Guide- Stone GW, Lindenfeld J, Abraham WT, et al. Transcatheter
lines. J Am Coll Cardiol 2014;63:2438-88. mitral-valve repair in patients with heart failure. N Engl
J Med 2018;379:2307-18.
Nkomo VT, Gardin JM, Skelton TN, et al. Burden of valvu-
lar heart diseases: a population-based study. Lancet Taniwaki M, Stefanini GG, Silber S, et al. RESOLUTE All-Com-
2006;368:1005-11. ers Investigators. 4-year clinical outcomes and predic-
tors of repeat revascularization in patients treated with
Nusca A, Bressi E, Colaiori I, et al. Antiplatelet therapy in val- new-generation drug-eluting stents: a report from the
vular and structural heart disease interventions. Cardio- RESOLUTE All-Comers trial (A Randomized Comparison
vasc Diagn Ther 2018;8:678-93. of a Zotarolimus-Eluting Stent with an Everolimus-Eluting
Stent for Percutaneous Coronary Intervention). J Am Coll
Otto CM, Nishimura RA, Bonow RO, et al. 2020 ACC/AHA Cardiol 2014;63:1617-25.
guideline for the management of patients with valvular
heart disease: a report of the American College of Cardiol- Tarantini G, Mojoli M, Varbella F, et al. Timing of oral P2Y12
ogy/American Heart Association Joint Committee on Clini- inhibitor administration in patients with non-ST-segment
cal Practice Guidelines. J Am Coll Cardiol 2021;77:e25-197. elevation acute coronary syndrome. J Am Coll Cardiol
2020;76:2450-9.
Otto CM, Prendergast B. Aortic-valve stenosis—from
patients at risk to severe valve obstruction. N Engl J Med Ten Berg J, Sibbing D, Rocca B, et al. Management of anti-
2014;371:744-56. thrombotic therapy in patients undergoing transcathe-
ter aortic valve implantation: a consensus document of
Rodés-Cabau J, Masson J-B, Welsh RC, et al. Aspirin versus
the ESC Working Group on Thrombosis and the European
aspirin plus clopidogrel as antithrombotic treatment
Association of Percutaneous Cardiovascular Interventions
following transcatheter aortic valve replacement with
(EAPCI), in collaboration with the ESC Council on Valvular
a balloon-expandable valve: the ARTE (aspirin versus
Heart Disease. Eur Heart J 2021;42:2265-9.
aspirin + clopidogrel following transcatheter aortic valve
implantation) randomized clinical trial. JACC Cardiovasc Valgimigli M, Bueno H, Byrne RA, et al. 2017 ESC focused
Interv 2017;10:1357-65. update on dual antiplatelet therapy in coronary artery dis-
ease developed in collaboration with EACTS. Eur Heart
Saad M, Meyer-Saraei R, de Waha-Thiele S, et al. Impact of
J 2018;39:213-60.
morphine treatment with and without metoclopramide
coadministration on ticagrelor-induced platelet inhibition Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus
in acute myocardial infarction: the randomized MonAMI clopidogrel in patients with acute coronary syndromes.
trial. Circulation 2020;141:1354-6. N Engl J Med 2009;361:1045-57.
Saito Y, Kobayashi Y. Triple therapy: A review of antithrom- Whitlow PL, Feldman T, Pedersen WR, et al. Acute and
botic treatment for patients with atrial fibrillation under- 12-month results with catheter-based mitral valve leaflet
going percutaneous coronary intervention. Journal of repair. J Am Coll Cardiol 2012;59:130-9.
Cardiology 2019;73:1-6.
Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus
Sarafoff N, Martischnig A, Wealer J, et al. Triple therapy with clopidogrel in patients with acute coronary syndromes.
aspirin, prasugrel, and vitamin K antagonists in patients N Engl J Med 2007;357:2001-15.
with drug-eluting stent implantation and an indication for
oral anticoagulation. J Am Coll Cardiol 2013;61:2060-6. Yasuda S, Kaikita K, Akao M, et al. Antithrombotic therapy
for atrial fibrillation with stable coronary disease. N Engl
Schömig A, Sarafoff N, Seyfarth M. Triple antithrombotic J Med 2019;381:1103-13.
management after stent implantation: when and how?
Heart 2009;95:1280-5. Yeh RW, Secemsky EA, Kereiakes DJ, et al. Development and
validation of a prediction rule for benefit and harm of dual
Schüpke S, Neumann F-J, Menichelli M, et al. Ticagrelor or antiplatelet therapy beyond 1 year after percutaneous cor-
prasugrel in patients with acute coronary syndromes. onary intervention. JAMA 2016;315:1735.
N Engl J Med 2019;381:1524-34.
You JJ, Singer DE, Howard PA, et al. Antithrombotic ther-
Sibbing D, Aradi D, Alexopoulos D, et al. Updated expert con- apy for atrial fibrillation: Antithrombotic Therapy and Pre-
sensus statement on platelet function and genetic testing vention of Thrombosis, 9th ed: American College of Chest
for guiding P2Y12 receptor inhibitor treatment in percuta- Physicians Evidence-Based Clinical Practice Guidelines.
neous coronary intervention. JACC: Cardiovascular Inter- Chest 2012;141:e531S-75S.
ventions 2019;12:1521-37.

PSAP 2022 Book 1 • Cardiology 110 Antithrombotic Therapy in Cardiac Interventions


Self-Assessment Questions
Questions 16–18 pertain to the following case. C. Start apixaban 5 mg twice daily, discontinue aspirin,
T.W. is a 58-year-old man with a history of heart failure with continue clopidogrel 75 mg daily.
preserved ejection fraction (HFpEF), hypertension, mitral D. Start apixaban 5 mg twice daily, discontinue aspirin
regurgitation (MR) and type 2 diabetes mellitus (T2DM). His and clopidogrel.
home drugs include losartan 100 mg daily, spironolactone
19. A 68-year-old man with a history of severe aortic steno-
25 mg daily, metformin 500 mg twice daily and sitagliptin
sis, atrial fibrillation, and hypertension is scheduled to
50 mg daily. Yesterday T.W. presented to the ED with chest
undergo a transcatheter aortic valve implantation (TAVI).
pressure and ECG demonstrated ST-elevations (STE). He was
His home drugs include losartan 100 mg daily, rosuvas-
diagnosed with STE myocardial infarction (STEMI) and under-
tatin 20 mg daily, metoprolol succinate 150 mg daily, and
went urgent percutaneous coronary intervention (PCI) and a
warfarin (INR goal 2.0–3.0). According to the most recent
drug-eluting stent was placed in the right coronary artery. 
European Society of Cardiology guidelines, which one of
16. Which one of the following antithrombotic regimens is the following antithrombotic therapies is best to recom-
best to recommend for T.W. after PCI for STEMI? mend for this patient post-TAVI?
A. Aspirin 81 mg daily plus prasugrel 10 mg daily for A. Warfarin (INR 2.0-3.0) indefinitely plus aspirin 81 mg
36 months daily for 6 months
B. Aspirin 81 mg daily plus ticagrelor 90 mg BID for B. Warfarin (INR 2.0-3.0) indefinitely
6 months C. Aspirin 81 mg daily and clopidogrel 75 mg daily for
C. Aspirin 81 mg daily plus ticagrelor 90 mg BID for 6 months, then warfarin INR goal 2.0 - 3.0)
12 months D. Rivaroxaban 10 mg daily and aspirin 81 mg daily
D. Aspirin 81 mg daily plus prasugrel 10 mg daily for indefinitely
6 months
20. Which one of the following patients undergoing PCI is
17. T.W. has been taking ticagrelor for 2 months. He calls the most likely to benefit from extended-DAPT duration
cardiology clinic requesting an alternative medication as beyond 12 months?
ticagrelor is too expensive. The cardiologist asks you to
A. 60-year-old man with a history of stage III chronic
provide a recommendation on how to switch to clopido-
kidney disease and peripheral arterial disease
grel. Which one of the following is best to recommend to
B. 48-year-old woman with hypertension
safely switch T.W. from ticagrelor to clopidogrel?
C. 50-year-old man, current smoker, with heart failure
A. Start clopidogrel 75 mg daily 24 hours after last with reduced ejection fraction and diabetes mellitus
dose of ticagrelor. D. 68-year-old man with a history of myocardial infarction
B. Start with a loading dose of clopidogrel 600 mg
21. A patient with a history of diabetes, ischemic stroke, and
24 hours after last dose of ticagrelor, then continue
hypertension presents with STEMI and is taken urgently
clopidogrel 75 mg daily thereafter.
to the catheterization laboratory. Because the patient
C. Stop ticagrelor, allow a washout period of 3 days,
was not loaded with an oral P2Y12 inhibitor, the interven-
then start clopidogrel 75 mg daily.
tionalist begins a cangrelor 30 mcg/kg bolus followed by
D. Stop ticagrelor, allow a washout period of 3 days,
4 mcg/kg/min infusion at the time of PCI. After comple-
then give a loading dose of clopidogrel 600 mg and
tion of cangrelor therapy, which one of the following is
continue clopidogrel 75 mg daily thereafter.
best to recommend to transition this patient to an oral
18. Two months later, T.W. is urgently seen in the cardiol- P2Y12 inhibitor?
ogy office for shortness of breath and intermittent pal-
A. Start clopidogrel 75 mg daily at time of cangrelor
pitations. Atrial fibrillation is noted on the ECG and oral
discontinuation.
anticoagulation is initiated. Which one of the following
B. Give a clopidogrel 600 mg loading dose immediately
adjustments to T.W.’s antithrombotic regimen is best to
before cangrelor discontinuation, then continue
recommend?
clopidogrel 75 mg daily thereafter.
A. Start warfarin (INR goal 2-3), continue aspirin 81 mg C. Give a prasugrel 60 mg loading dose 60 minutes
daily and clopidogrel 75 mg daily. before cangrelor discontinuation, then continue
B. Start warfarin (INR goal 2-3), discontinue aspirin, prasugrel 10 mg daily thereafter.
continue clopidogrel 75 mg daily. D. Give a ticagrelor 180 mg loading dose as soon as
possible before cangrelor discontinuation, then
continue ticagrelor 90 mg twice daily thereafter.

PSAP 2022 Book 1 • Cardiology 111 Antithrombotic Therapy in Cardiac Interventions


22. A 68-year-old man has a medical history of severe aortic Questions 26 and 27 pertain to the following case.

stenosis in the setting of a bicuspid aortic valve, hyper- W.B. is a 67-year-old man who underwent placement of a DES
tension, and hyperlipidemia. He undergoes an elective to his left circumflex coronary artery 5 days ago. He is pre-
TAVI and is admitted to the cardiac stepdown unit for scribed aspirin and clopidogrel upon discharge. 
observation after the procedure. According to the Euro- 26. W.B. asks you why he is being prescribed clopidogrel
pean Society of Cardiology consensus statement, which and aspirin. This dual antiplatelet therapy will most
one of the following is best to recommend as the anti- likely reduce W.B.’s risk of which one of the following
thrombotic regimen for this patient after successful complications?
TAVI?
A. In-stent restenosis
A. Aspirin 81 mg daily B. Early stent thrombosis
B. Aspirin 81 mg daily plus clopidogrel 75 mg daily C. Very late stent thrombosis
C. Warfarin with goal INR of 2-3 D. Stroke
D. Rivaroxaban 10 mg daily plus aspirin 81 mg daily
27. Three years later, W.B. is admitted for progressive
23. A 75-year-old woman has a medical history of stable angina, and he undergoes a coronary angiogram. The
coronary artery disease status post-PCI with place- angiogram reveals significant in-stent restenosis (ISR),
ment of a drug-eluting stent (DES) to left main and and he undergoes orbital atherectomy with repeat stent-
proximal left anterior descending artery (2 months ing to his left circumflex coronary artery. Which one of
ago), hypertension, T2DM, and GERD. The patient pres- the following is most likely associated with W.B.’s ISR?
ents to the cardiology service after elective TAVI. Her
A. Poor adherence with DAPT
home drugs include aspirin 81 mg daily, atorvastatin
B. Coronary artery hyperplasia
40 mg daily, carvedilol 6.25 mg BID, clopidogrel 75 mg
C. Stent underexpansion
daily, lisinopril 10 mg daily, metformin 500 mg BID, and
D. Impaired re-endothelialization 
pantoprazole 40 mg daily. Which one of the following is
best to recommend for this patient after her TAVI? 28. Emergency Medical Services responds to the local gro-
cery store to assist a patient who is diaphoretic with
A. Aspirin 81 mg daily indefinitely, discontinue
crushing chest pain. Morphine 2 mg IV for chest pain is
clopidogrel
administered in the ambulance on the way to the hos-
B. Aspirin 81 mg daily plus clopidogrel 75 mg daily for
pital. Upon arrival, the patient is diagnosed with an
4 months, followed by aspirin indefinitely
ST-elevation MI, given 180 mg of ticagrelor and is taken
C. Rivaroxaban 10 mg daily plus clopidogrel 75 mg
to the catheterization lab for PCI 10 minutes later. Due to
daily, discontinue aspirin
the interaction between morphine and ticagrelor, which
D. Aspirin 81 mg daily plus clopidogrel daily for
one of the following laboratory abnormalities is mostly
10 months, followed by clopidogrel indefinitely
likely to be seen after this patient’s PCI?
24. A patient with a history of recent venous thromboembo-
A. Reduced troponin-I, reduced platelet reactivity
lism on apixaban is undergoing TAVI. Which one of the
B. Increased troponin-I, increased platelet reactivity
following is best to recommend for this patient after
C. Increased platelet reactivity, increased lactate
TAVI?
D. Reduced platelet reactivity, increased lactate
A. Apixaban 5 mg PO BID
29. A 49-year-old man presents to the ED and is diagnosed
B. Apixaban 5 mg PO BID plus aspirin 81 mg daily
with a non ST-elevation MI. He has no contributory med-
C. Apixaban 5 mg PO BID plus clopidogrel 75 mg daily
ical history, but his BP on presentation is 156/74 mm Hg
D. Apixaban 5 mg PO BID plus ticagrelor 90 mg BID
and A1C is found to be 6.8%. His PRECISE-DAPT score is
25. A 76-year-old man with a history of heart failure with 14. The team is planning to take him for coronary angiog-
reduced ejection fraction and hypertension is under- raphy and PCI with stent placement. Which one of the fol-
going Mitraclip placement. After Mitraclip placement, lowing P2Y12 inhibitor administration strategies is best to
which one of the following is best to recommend for this recommend for this patient?
patient?
A. Prasugrel 60 mg x 1 dose before coronary
A. Aspirin 81 mg daily angiography
B. Aspirin 81 mg daily plus clopidogrel 75 mg daily B. Clopidogrel 600 mg x 1 dose 2 hours after PCI
C. Aspirin 81 mg daily plus ticagrelor 90 mg BID C. Ticagrelor 180 mg x 1 dose before coronary
D. Aspirin 81 mg daily plus prasugrel 10 mg daily angiography
D. Clopidogrel 300 mg x 1 dose immediately after PCI

PSAP 2022 Book 1 • Cardiology 112 Antithrombotic Therapy in Cardiac Interventions


30. A 59-year-old man is admitted to the coronary care unit for
NSTE-ACS. His medical history includes atrial fibrillation,
familial hypercholesterolemia, coronary artery disease
status post-multiple PCIs to left circumflex artery (2017),
right coronary artery (2019), hypertension, chronic kidney
disease stage III, and T2DM. His home drugs include apix-
aban 5 mg BID, aspirin 81 mg daily, atorvastatin 80 mg
daily, evolocumab 420 mg SQ every 2 weeks, lisinopril 20
mg daily, metoprolol succinate 100 mg daily. The patient
undergoes PCI to his left main and proximal left anterior
descending artery and is started on DAPT. Which one of
the following antithrombotic therapy regimens is best to
recommend for this patient?
A. Apixaban 5 mg BID plus aspirin 81 mg daily plus
clopidogrel 75 mg daily for 30 days, then apixaban
5 mg BID and clopidogrel 75 mg daily for to
12 months
B. Apixaban 5 mg BID plus aspirin 81 mg daily plus
clopidogrel 75 mg daily for 12 months, then
apixaban monotherapy
C. Apixaban 5 mg BID plus clopidogrel 75 mg daily for
12 months
D. Apixaban 5 mg BID plus prasugrel 10 mg daily for
12 months

PSAP 2022 Book 1 • Cardiology 113 Antithrombotic Therapy in Cardiac Interventions


Learner Chapter Evaluation: Antithrombotic Therapy in Cardiac Interventions

As you take the posttest for this chapter, also evaluate the 25. The learning assessment activities used in the chapter
material’s quality and usefulness, as well as the achievement were effective.
of learning objectives. Rate each item using this 5-point scale: 26. The chapter was effective overall.

• Strongly agree 27. The activity met the stated learning objectives.
• Agree 28. If any objectives were not met, please list them here.
• Neutral
• Disagree OTHER COMMENTS
• Strongly disagree
29. Please provide any specific comments related to any
16. The content of the chapter met my educational needs. perceptions of bias, promotion, or advertisement of
commercial products.
17. The content of the chapter satisfied my expectations.
30. Please expand on any of your above responses, and/or
18. The author presented the chapter content effectively.
provide any additional comments regarding this chapter:
19. The content of the chapter was relevant to my practice
and presented at the appropriate depth and scope.
Questions 31–33 apply to the entire learning module.
20. The content of the chapter was objective and balanced.
31. How long did it take you to read the instructional materi-
21. The content of the chapter is free of bias, promotion, and
als in this module?
advertisement of commercial products.
32. How long did it take you to read and answer the assess-
22. The content of the chapter was useful to me.
ment questions in this module?
23. The teaching and learning methods used in the chapter
33. Please provide any additional comments you may have
were effective.
regarding this module:
24. The active learning methods used in the chapter were
effective.

PSAP 2022 Book 1 • Cardiology 114 Antithrombotic Therapy in Cardiac Interventions


Cardiology III
Cardiology III Panel

Series Editors: Joel Peterson, Pharm.D., BCPS


Cynthia A. Sanoski, Pharm.D., FCCP, BCPS Clinical Pharmacist
PGY-1 Residency Program Director
Department Chair
Department of Pharmacy
Associate Professor of Pharmacy Practice
Hennepin Healthcare
Thomas Jefferson University
Minneapolis, Minnesota
Philadelphia, Pennsylvania
Daniel M. Witt, Pharm.D., FCCP, BCPS
Professor and Chair Non-Statin Therapy for Dyslipidemia
Department of Pharmacotherapy Authors
Assistant Dean of Clinical Affairs
Nicholas W. Carris, Pharm.D., BCPS
University of Utah College of Pharmacy
Salt Lake City, Utah Associate Professor
Department of Pharmacotherapeutics and Clinical
Faculty Panel Chair: Research
USF Health Taneja College of Pharmacy
Craig J. Beavers, Pharm.D., FCCP, FACC, FAHA,
Tampa, Florida
BCCP, BCPS-AQ Cardiology, CACP
Director of Cardiovascular Services Kevin Cowart, Pharm.D., MPH, BCACP, CDCES
Baptist Health Paducah Assistant Professor
Cardiovascular Clinical Pharmacist Department of Pharmacotherapeu-
Department of Pharmacy Services tics and Clinical Research
UK Healthcare USF Health Taneja College of Pharmacy
Assistant Adjunct Professor Tampa, Florida
Department of Pharmacy Practice and Science
University of Kentucky College of Pharmacy Reviewers
Paducah, Kentucky John Bucheit, Pharm.D., BCACP, CDCES
Associate Professor
Department of Pharmacotherapy and Outcomes Science
Management of Atrial Fibrillation VCU School of Pharmacy
Author Richmond, Virginia
Amy L. Lehnert, Pharm.D., BCPS, BCCP Gregory Castelli, Pharm.D., BCPS, BC-ADM
Cardiology Clinical Pharmacy Specialist Clinical Pharmacist
Roper Hospital Director, PGY1 Residency
Charleston, South Carolina Department of Medical Education
UPMC St. Margaret
Reviewers Adjunct Assistant Professor
Ashley Schenk, Pharm.D., BCPS, BCCP Department of Pharmacy and Therapeutics
Cardiology Clinical Pharmacist University of Pittsburgh
Department of Pharmacy Pittsburgh, Pennsylvania
UK HealthCare Eugene N. Bush, Ph.D., BCPS, BCCCP
Assistant Adjunct Professor Clinical Staff Pharmacist
Department of Pharmacy Practice and Science Vista Medical Center East
University of Kentucky College of Pharmacy Waukegan, Illinois
Lexington, Kentucky
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
Consultancies: Anastasia Armbruster (ACC/AHA Joint Committee Clinical Practice Guidelines); Jessica Bente (ACCP); Allison
Burnett: Consultancies (Anticoagulation Forum; National Certification Board for Anticoagulation Providers; Global Strategies);
Jessie L. Dunne (Pulmonary Hypertension Association); Nancy M. Nix (Pharmacosmos); Daniel M. Riche (Novo Nordisk; Merck;
AstraZeneca); Ralph J. Riello III (AstraZeneca; Johnson & Johnson); Sara Vazquez (Anticoagulation Forum);

Stock Ownership:

Royalties: Allison Burnett (Wolters-Kluwer); Daniel M. Riche (McGraw-Hill); Honoraria (Merck); Sara Vazquez (UpToDate)

Grants: Anastasia Armbruster (ACCP); Jessica Bente (New Jersey Society of Health-System Pharmacists); Gregory Castelli
(American Board of Family Medicine); Kevin Cowart (University of South Florida Taneja College of Pharmacy); Ralph J. Riello
III (AstraZeneca)

Honoraria: Anastasia Armbruster (AstraZeneca); John Bucheit (Association of Diabetes Care & Education Specialists); Nancy M.
Nix (Coherus; Pfizer; AstraZeneca); Ralph J. Riello III (Alexion; AstraZeneca; Janssen)

Other:

Nothing to disclose: Katherine Aymond, Eugene N. Bush, Nicholas W. Carris, Stephanie Dwyer Kaluzna, Lindsey Federle, Beth-
any A. Ford, Stormi E. Gale, Jaclynne R. Gowen, Glenn Herrington, Christine Ji, Amy L. Lehnert, Nilam R. Naik, Brenda Pahl, Joel
J. Peterson, A. Joshua Roberts, Kelly M. Rudd, Ashley Schenk, Susan M. Smith, Elisabeth M. Wang, Toby C. Trujillo, Barbara Wig-
gins, Leslie Wooten, Eman El Sayed Younis

ROLE OF BPS: The Board of Pharmacy Specialties (BPS) is an autonomous division of the American Pharmacists Association (APhA).
To maintain its strict, independent standards for certification, BPS does NOT endorse or provide review information, prepara-
tory courses, or study guides for Board Certification Examinations. The Board, through its specialty councils, is responsible for
specialty examination content, administration, scoring, and all other aspects of its certification programs. BPS is totally separate
and distinct from ACCP. PSAP has been approved by BPS for use in BCPS recertification. Information about the BPS recertification
process is available online.

Questions regarding BPS specialty recertification should be directed to:

Board of Pharmacy Specialties


2215 Constitution Avenue NW
Washington, DC 20037
(202) 429-7591
Continuing Pharmacy Education
and Recertification Instructions
Continuing Pharmacy Education (CPE) Credit: The American College of Clinical Pharmacy is accredited by the
Accreditation Council for Pharmacy Education (ACPE) as a provider of CPE.

PSAP Target Audience: The target audience for PSAP 2022 Book 1 (Cardiology) is pharmacotherapy specialists and advanced
level clinical pharmacists encountering diverse cardiovascular patient populations.

Available CPE credits: Purchasers who successfully complete all posttests for PSAP 2022 Book 1 (Cardiology) can earn 21.5 contact
hours of CPE credit. The universal activity numbers are as follows: Cardiology I – 0217-0000-22-002-H01-P, 5.0 contact hours;
Cardiology II – 0217-0000-22-003-H01-P, 6.0 contact hours; Cardiology III – 0217-0000-22-004-H01-P, 5.0 contact hours; and
Cardiology IV – 0217-0000-22-005-H01-P, 5.5 contact hours. You may complete one or all available modules for credit. Tests may
not be submitted more than one time.

TO EARN CPE CREDITS FROM THIS PSAP BOOK

Posttest access: Go to www.accp.com and sign in with your e-mail address and password. Technical support is available from 8 a.m.
to 5 p.m. (Central) weekdays by calling (913) 492-3311. PSAP products are listed under My Products on your My Account page.

BCPS test deadline: 11:59 p.m. (Central) on July 15, 2022.


ACPE test deadline: 11:59 p.m. (Central) on January 18, 2025.

BCPS Recertification Credit: To receive BCPS recertification CPE credit, a PSAP posttest must be submitted within
the 6-month period after the book’s release (see above). Only completed tests are eligible for credit; no partial or
incomplete tests will be processed. You may complete one or all available modules for credit. Tests may not be
submitted more than one time.

The passing point to earn BCPS recertification credit is based on an expert analysis of the assessment items in each posttest
module. Any posttest submitted before the BCPS test deadline that meets this passing point will earn BCPS recertification
credits. These credits will be assigned as of the date of test submission and reported within 48 hours to BPS. For statements of
recertification credit, visit www.bpsweb.org.

Questions regarding the number of hours required for BCPS recertification should be directed to BPS at (202) 429-7591 or
www.bpsweb.org. The ACCP Recertification Dashboard is a free online tool that can track recertification credits as they are
earned through ACCP and schedule new opportunities for credits from upcoming ACCP professional development programs.

ACPE CPE Credit: To receive ACPE CPE credit for a PSAP module, a posttest must be submitted within 3 years after the book’s
release (see above). Only completed tests are eligible for credit; no partial or incomplete tests will be processed. You may
complete one or all available modules for credit. Tests may not be submitted more than one time.

Any posttest submitted before the ACPE deadline that scores 50% or greater will be awarded the appropriate CPE. These
credits will be assigned as of the date of test submission and reported within 48 hours. For statements of CPE credit, visit
www.mycpemonitor.net.

Posttest answers: The explained answers—with rationale and supporting references—will be posted s weeks after the BCPS
test deadline and will be available to anyone who has either (1) submitted a posttest or (2) waived the right to receive credit from
a posttest (see below). Go to www.accp.com and sign in with your e-mail address and password. Click the PSAP book on your
My Account page and you will see a link to the explained answers.

Test Waivers: To access the explained answers without submitting a posttest, sign in to your My Account page, select the PSAP
book, and click on the waiver link for that module. By completing the waiver form for a module, you waive the opportunity to
receive CPE credit for that module. After you submit a waiver, you will see a link to the PDF file that contains the answers for the
module you waived. Answers will be available starting 2 weeks after the BCPS test deadline.
Management of Atrial Fibrillation
By Amy L. Lehnert, Pharm.D., BCPS, BCCP

Reviewed by Ashley Schenk, Pharm.D., BCPS, BCCP; and Joel Peterson, Pharm.D., BCPS

LEARNING OBJECTIVES

1. Evaluate various types of atrial fibrillation (AF), clinical features, and risk factors according to current literature and
guideline recommendations.
2. Justify the place in therapy of oral anticoagulants in AF management.
3. Evaluate differences between rate- and rhythm-control strategies in AF management.
4. Assess nonpharmacologic therapies for stroke prevention and rhythm control in AF, together with clinical considerations
surrounding anticoagulation, complications, and contraindications to these therapies.
5. Develop an appropriate treatment plan for AF management in special patient populations.

INTRODUCTION
ABBREVIATIONS IN THIS CHAPTER
Atrial fibrillation (AF) is the most common cardiac arrhythmia in
AAD Antiarrhythmic drug
adults worldwide. Complications from AF are associated with sub-
AC Anticoagulation
stantial morbidity and mortality and pose a significant burden on
ACS Acute coronary syndrome
patients, physicians, and health care systems (Hindricks 2021). The
AF Atrial fibrillation
best strategies for managing AF and preventing complications have
AV Atrioventricular
been debated for decades, and newer technologies have evolved to
CAD Coronary artery disease
add to the arsenal of detection and treatment modalities for this dis-
CV Cardiovascular
ease. This chapter discusses the current pharmacologic and non-
DCCV Direct current cardioversion
pharmacologic management strategies for AF and how they are
DOAC Direct oral anticoagulant implemented in practice.
HF Heart failure
HFrEF Heart failure with reduced ejection Background
fraction
Currently, AF affects around 43.6 million people worldwide and more
LA Left atrium
than 3 million Americans (Hindricks 2021; Chung 2020). As patient
LAA Left atrial appendage
life expectancy increases and AF detection methods improve, these
LMWH Low-molecular-weight heparin
numbers are expected to double over the next 25 years (January
LV Left ventricular
2014). The prevalence of AF increases with age, from 0.5% in indi-
MI Myocardial infarction viduals 50–59 years of age to greater than 33% in those older than
OAC Oral anticoagulation 80 (Hindricks 2021). Complications of AF include frequent hospital-
PV Pulmonary vein izations, thromboembolic events, hemodynamic abnormalities, and
TEE Transesophageal echocardiogram decreased patient quality of life. In the United States, patients with
TIA Transient ischemic attack AF are hospitalized twice as often as patients without AF and are 3
UFH Unfractionated heparin times more likely to have several hospital admissions. Stroke risk is
increased by 5-fold in patients with AF, with AF-related stroke typically
Table of other common abbreviations.
more severe than non–AF-related stroke. Finally, a 3-fold increased
risk of heart failure (HF) and 2-fold increased risk of both dementia
and mortality have been associated with AF (January 2014).

PSAP 2022 Book 1 • Cardiology 121 Management of Atrial Fibrillation


Clinical Features include fatigue, palpitations, dyspnea, hypotension, syncope,
Atrial fibrillation is caused by a lack of organized atrial depo- or HF, with fatigue being the most common. Hemodynamic
larization, leading to chaotic and disorganized atrial activity. disturbances from AF may be caused by abnormal ventricu-
Characteristic ECG findings include irregular R-R intervals, lar conduction rates (either too fast or too slow), loss of atrial
an absence of distinct repeating P waves, and irregular contraction (i.e., loss of atrial “kick”), inconsistent ventricu-
atrial activity. Clinical symptoms of AF vary, but 50%–87% lar filling time, and increased adrenergic tone (January 2014).
of patients are initially asymptomatic. Other symptoms may
DEFINITIONS
Table 1 summarizes the different types of AF that can be
BASELINE KNOWLEDGE STATEMENTS described on the basis of presentation, duration, and sponta-
neous termination of episodes. Over time, AF episodes often
Readers of this chapter are presumed to be familiar
with the following: increase in duration and frequency and in patients experienc-
ing more than one type of AF – for example, with paroxys-
• General knowledge of the pathophysiology that mal and persistent AF, the more common type should be used
leads to atrial fibrillation (AF)
for classification (January 2014). Implantable cardiac elec-
• Baseline familiarity with ECGs
tronic devices (i.e., implantable loop recorders, pacemakers,
• General drug knowledge of the various oral and defibrillators) offer enhanced reporting capabilities of AF epi-
intravenous pharmacologic agents used to treat AF
sode duration, rate, and frequency; this collection of informa-
• Familiarity with guideline-recommended risk
assessment modalities and a general understand- tion is often called the “AF burden.”
ing of the clinical sequelae of AF, namely stroke Atrial fibrillation can also be described with respect to
and systemic embolism the presence of certain types of valvular disease and valvu-
lar intervention. Differentiating between “valvular” and “non-
Table of common laboratory reference values
valvular” AF has been confusing for clinicians for many years
because of varying definitions between guidelines and clinical
ADDITIONAL READINGS trials. The 2019 American Heart Association/American College
of Cardiology/Heart Rhythm Society (AHA/ACC/HRS) focused
The following free resources have additional back-
update of the 2014 AHA/ACC/HRS guideline for the treatment
ground information on this topic:
of patients with AF clarified this terminology (see Table 1). Sup-
• Hindricks G, Potpara T, Dagres N, et al. 2020 ESC port for this distinction in terminology stems from the direct
guidelines for the diagnosis and management of
atrial fibrillation developed in collaboration with the oral anticoagulant (DOAC) AF clinical trials, in which about 20%
European Association for Cardio-Thoracic Surgery of patients were enrolled with various valvular defects, includ-
(EACTS). Eur Heart J 2021;42:373-498. ing mild mitral stenosis, mitral regurgitation, aortic stenosis,
• January CT, Wann LS, Calkins H, et al. 2019 AHA/ aortic regurgitation, and tricuspid regurgitation, together with
ACC/HRS focused update of the 2014 AHA/ACC/ some patients with valve repair, valvuloplasty, and biopros-
HRS guideline for the management of patients with thetic valves. Meta-analyses suggest that among patients
atrial fibrillation: a report of the American College with AF and these valvular disorders, DOACs better reduce
of Cardiology/American Heart Association Task
stroke and systemic embolism than warfarin, but bleeding risk
Force on Clinical Practice Guidelines and the Heart
Rhythm Society in Collaboration with the Society varies among the different DOACs (January 2019).
of Thoracic Surgeons [published correction
appears in Circulation 2019;140:e285]. Circulation MECHANISMS
2019;140:e125-e151. Structural and/or electrophysiologic cardiac abnormalities
• Calkins H, Hindricks G, Cappato R, et al. 2017 HRS/ can alter atrial tissue, promoting abnormal impulse forma-
EHRA/ECAS/APHRS/SOLAECE expert consensus
tion and/or propagation, causing AF. These abnormalities may
statement on catheter and surgical ablation of
be precipitated by a wide variety of pathophysiologic mecha-
atrial fibrillation. Heart Rhythm 2017;14:e275-e444.
nisms, many of which are incompletely or poorly understood.
• January CT, Wann LS, Alpert JS, et al. 2014 AHA/ Ultimately, AF reflects a final common phenotype for a complex
ACC/HRS guideline for the management of patients
with atrial fibrillation: a report of the American Col- interplay of triggers, substrates, and physiologic processes,
lege of Cardiology/American Heart Association often making it difficult for clinicians to manage (January
Task Force on Practice Guidelines and the Heart 2014). Figure 1 summarizes potential AF mechanisms.
Rhythm Society [published correction appears in J
Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol Pathophysiology
2014;64:e1-e76.
For AF to occur, an initiation trigger and maintenance sub-
strate are required. Many hypotheses have surrounded the

PSAP 2022 Book 1 • Cardiology 122 Management of Atrial Fibrillation


Table 1. Classification of AF

Term Definition

Paroxysmal AF that terminates spontaneously or with intervention within 7 days of onset


Episodes may recur with variable frequency

Persistent Continuous AF that is sustained beyond 7 days, including episodes terminated by cardioversion (drugs or
electrical cardioversion) at or beyond 7 days

Longstanding Continuous AF beyond 12 mo when the patient and physician have decided to adopt a rhythm-control
persistent strategy

Permanent AF that is accepted by the patient and physician, and no further attempts to restore/maintain sinus rhythm
will be made. Acceptance of AF represents a therapeutic attitude on the part of the patient and clinician
rather than an inherent pathophysiologic attribute of AF, and the term should not be used in the context of
a rhythm-control strategy with AAD therapy or AF ablation. Acceptance of AF may change as symptoms,
efficacy of interventions, and patient/clinician preferences evolve; should a rhythm-control strategy be
adopted, AF will be reclassified as “longstanding persistent AF”

Nonvalvular AF AF in the absence of moderate to severe mitral stenosis or a mechanical heart valve. Nonvalvular AF does not
imply the absence of valvular heart disease

Valvular AF AF in the setting of moderate to severe mitral stenosis or a mechanical heart valve

Terminology That Should Be Abandoned

Lone AF A historical descriptor that has been variably applied to younger individuals without clinical or
echocardiographic evidence of cardiopulmonary disease, HTN, or diabetesa

Chronic AF Has variable definitions and should not be used to describe populations of patients with AF

a
Increasing knowledge about the pathophysiology of AF shows that in every patient, a cause is present. Hence, this term is potentially
confusing and should be abandoned.
AAD = antiarrhythmic drug; AF = atrial fibrillation; HTN = hypertension.
Information from: Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of atrial fibrillation
developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J 2021;42:373-498;
January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/ACC/HRS guideline for the
management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task
Force on Clinical Practice Guidelines and the Heart Rhythm Society in Collaboration with the Society of Thoracic Surgeons
[published correction appears in Circulation 2019;140:e285]. Circulation 2019;140:e125-e151; January CT, Wann LS, Alpert JS, et al.
2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of
Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society [published correction
appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

pathophysiology of both AF initiation and maintenance; how- refractoriness, several rapidly firing foci in response to cardiac
ever, for most patients, this relationship is likely multifactorial ganglionic plexi activity, and several rotors or spiral wave reen-
and extremely complex (January 2014). trant circuits. Several AF therapies have been developed on the
Atrial fibrillation is typically triggered by rapidly firing ecto- basis of these proposed mechanisms, namely surgical maze
pic focal discharges, which are abnormal electrical impulses and catheter ablation procedures (January 2014).
originating from atrial tissue. These impulses lead to disorga-
nized atrial electrical activity, with subsequent lack of cohe- Risk Factors
sive atrial depolarization and mechanical contraction. The Like many chronic conditions, AF is highly associated with
impulses most commonly originate from the left atrial (LA) many common disease states, underscoring the importance
myocardial sleeves extending into the pulmonary veins (PVs), of lifestyle modifications as a key aspect of AF prevention
making this area one of the primary targets in catheter ablation and treatment. Box 1 summarizes the many nonmodifiable
procedures. Another potential trigger for AF is abnormal cal- and modifiable risk factors that have been recognized in the
cium handling in which calcium “leaks” from the sarcoplasmic development of AF. Some of these conditions can lead to pro-
reticulum during diastole, causing delayed afterdepolarizations gressive increases in atrial enlargement, LA fibrosis, auto-
(January 2014). Proposed theories for AF maintenance include nomic dysfunction, and left ventricular (LV) hypertrophy,
reentrant wavelets caused by heterogeneous conduction and further increasing susceptibility to AF (Ponamgi 2021).

PSAP 2022 Book 1 • Cardiology 123 Management of Atrial Fibrillation


Inflammation

Oxidative Stress Atrial Electrical


Atrial Structural
Abnormalities:
Abnormalities:
↑ Heterogeneity
Fibrosis
↓ Conduction
Dilation
↓ AP duration/refractoriness
Ischemia
↑ Automaticity
Infiltration
Abnormal intracellular Ca++
Hypertrophy
handling

AF

Genetic RAAS activation


Variants:
Autonomic
Channelopathy nervous system
Cardiomyopathy activation

Extracardiac Risk
Factors:

HTN
Obesity
Sleep apnea
Hyperthyroidism
Alcohol/drugs

Figure 1. Mechanisms of atrial fibrillation.


AF = atrial fibrillation; AP = action potential; Ca++ = ionized calcium; HTN = hypertension; RAAS = renin-angiotensin-aldosterone system.
Information from: January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial
fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the
Heart Rhythm Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

Box 1. Potential Modifiable and Nonmodifiable Risk Factors for AF


Nonreversible Risk Factors • Chronic kidney disease
• AF risk genotypes • Chronic obstructive pulmonary disease
• Family history of AF • Coronary artery disease
• Height • Diabetes
• Older age • Heart failure/cardiomyopathy
Reversible Risk Factors • Hypertension
• Acute illness/surgery • Hyperthyroidism
• Alcohol/drugs/smoking • Inflammatory diseases
• Cardiac surgery • Obesity
• Chronic endurance training • Sleep apnea
• Valvular heart disease
AF = atrial fibrillation.
Information from: Ponamgi SP, Siontis KC, Rushlow DR, et al. Screening and management of atrial fibrillation in primary care. BMJ
2021;373:n379.

PSAP 2022 Book 1 • Cardiology 124 Management of Atrial Fibrillation


DIAGNOSTIC CRITERIA Thus, risk assessment tools have been developed and vali-
Because most AF is asymptomatic, it is ultimately diagnosed dated to help stratify both thrombotic and bleeding risk.
by ECG, with episodes lasting at least 30 seconds indicative The CHA 2DS2-VASc risk scoring system is recommended
of clinical AF (Hindricks 2021; January 2014). Physical exam- by the current guidelines to estimate annual stroke risk
ination findings may include an irregular pulse, irregular jugu- and guide decisions surrounding anticoagulant therapy
lar venous pulsations, or variations in the first or fourth heart (Table 2). Although the CHA 2DS2-VASc performs only mod-
sound. Initial diagnosis of AF involves characterizing pattern estly in predicting stroke in high-risk patients, it consistently
type (i.e., paroxysmal, persistent, longstanding persistent,
permanent), identifying potential causes, defining associ-
ated comorbidities, and assessing stroke risk. In addition, an
Table 2. CHA 2DS2-VASc Scoring System
echocardiogram should be performed to identify underlying
structural heart disease and assess cardiac function. Serum
Step 1. Identify qualifying criteria
electrolytes and CBC, together with thyroid, renal, and hepatic
Point Value Criteria
function testing, should also be assessed (January 2014).
1 C – Congestive HF/LV dysfunction
CLINICAL MANAGEMENT OF AF 1 H – HTNa
Once diagnosed, ideal AF management should involve a coor- 2 A 2 – Age ≥ 75
dinated multidisciplinary team approach to deliver individu-
1 D – Diabetesb
alized patient care. Goals of care should center on reducing
stroke risk, improving symptoms, and treating comorbidi- 2 S2 – Stroke/TIA/TEc
ties while recognizing that patient treatment plans are sub- 1 V – Vascular diseased
ject to change over time with development of new symptoms,
1 A – Age 65–74
disease progression, and introduction of new treatment
modalities. The 2020 European Society of Cardiology (ESC) 1 Sc – Female sex
guidelines describe this approach with the “Atrial fibrillation Step 2. Determine annual stroke risk
Better Care” (ABC) holistic pathway (“A” Anticoagulation/
Point Total Adjusted Stroke Rate (% per year)
Avoid stroke; “B” Better symptom management; “C” Cardio-
1 1.3
vascular and Comorbidity optimization) (Hindricks 2021). In
several studies, implementation of the ABC pathway has sig- 2 2.2
nificantly reduced the risk of all-cause death, the composite 3 3.2
outcome of stroke/major bleeding/cardiovascular (CV) death
and first hospitalization, CV event rates, and health-related 4 4.0
costs compared with usual care (Pastori 2019a, 2019b; Yoon 5 6.7
2019; Proietti 2018; Lip 2017).
6 9.8

Stroke Prevention 7 9.6

Stroke is one of the most devastating potential complica- 8 6.7


tions of AF, with the left atrial appendage (LAA) being one 9 15.2
of the most common sites of thrombus formation and sub-
sequent thromboembolization. Compared with patients in a
Resting blood pressure > 140/90 mm Hg on at least two occa-
normal sinus rhythm, the risk of stroke is increased by 5-fold sions or current antihypertensive pharmacologic treatment.
in nonvalvular AF and 20-fold in AF in the setting of mitral b
Fasting glucose > 125 mg/dL or treatment with oral
stenosis. Atrial fibrillation–related stroke is also associated hypoglycemic agent and/or insulin.
with greater severity of disability, risk of recurrent stroke,
c
Includes any history of cerebral ischemia.
and mortality than non–AF-related stroke (January 2014;
d
Prior MI, PAD, or aortic plaque.
HF = heart failure; LV = left ventricle; MI = myocardial
Lin 1996).
infarction; PAD = peripheral arterial disease;
TE = thromboembolism; TIA = transient ischemic attack.
Risk Assessment
Information from: January CT, Wann LS, Alpert JS, et al.
Although oral anticoagulation (OAC) is one of the cornerstones 2014 AHA/ACC/HRS guideline for the management of
of therapy in successfully reducing the risk of AF-related patients with atrial fibrillation: a report of the American
stroke, it is also associated with an increased risk of bleeding. College of Cardiology/American Heart Association Task
Force on Practice Guidelines and the Heart Rhythm
In addition, although AF increases stroke risk, this risk is not
Society [published correction appears in J Am Coll Cardiol
homogeneous and may vary significantly between patients 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.
depending on differences in risk factors and comorbidities.

PSAP 2022 Book 1 • Cardiology 125 Management of Atrial Fibrillation


identifies and differentiates patients at low to moderate includes an assessment of both modifiable and nonmodifi-
risk, which significantly helps in determining which patients able risk factors, with a score of 3 or greater indicating poten-
should receive anticoagulant therapy. The CHA 2DS2-VASc tially “high” bleeding risk. Several of the bleeding risk factors
score component of female sex has elicited many ques- included in the HAS-BLED scoring tool are also described
tions from practicing clinicians and may be more easily in the CHA 2DS2-VASc scoring tool as stroke risk factors. As
described as an age-dependent stroke risk modifier than such, a high bleeding risk score should not necessarily lead
a risk factor (Hindricks 2021). For example, observational to withholding anticoagulation because these patients are
data show that women with no other stroke risk factors also likely to have a higher risk of stroke, and the net clini-
(CHA 2DS2-VASc score 1) have a low stroke risk, similar to men cal benefit with continued anticoagulation is greater. Rather,
with a CHA 2DS2-VASc score of 0 (Nielsen 2020). However, in a high bleeding risk score should prompt focus on correction
the presence of more than one non-sex stroke risk factor, of modifiable bleeding risk factors and closer monitoring of
women with AF consistently have a higher stroke risk than nonmodifiable risk factors. Of note, a history of falls is not
men (Marzona 2018). Thus, not considering the sex compo- included in the HAS-BLED score because it is not an indepen-
nent in the CHA 2DS2-VASc scoring tool would significantly dent predictor of bleeding in patients receiving anticoagula-
underestimate stroke risk in women with AF. tion (Hindricks 2021; January 2014).
When initiating anticoagulant therapy for AF, the poten-
Anticoagulant Therapy
tial risk of bleeding should also be assessed. The HAS-BLED
score is recommended by the current guidelines to help iden- Oral anticoagulation therapy is the first-line treatment for
tify patients who are at increased risk of bleeding and may stroke prevention in AF and includes the DOACs apixaban,
require closer monitoring (Table 3). The HAS-BLED score rivaroxaban, edoxaban, and dabigatran, together with warfa-
rin. Anticoagulation therapy helps prevent strokes and sys-
temic emboli among patients in AF, largely by reducing the
formation of thrombi in the LA or LAA. However, because anti-
Table 3. HAS-BLED Scoring System
coagulation increases the risk of bleeding, its benefits must
Point be weighed against the risk of harm. Current guidelines rec-
Value Criteria ommend use of the CHA 2DS2-VASc score to assess stroke risk,
1 H – HTN (uncontrolled, SBP > 160 mm Hg) with anticoagulation initiation if CHA 2DS2-VASc is 1 or greater
in men or 2 or greater in women (IIb, C-LD). The recommen-
1a A – Abnormal renal and/or hepatic functionb
dation for anticoagulation strengthens to I-A if the CHA 2DS2-
1 S – Stroke (ischemic or hemorrhagicc) VASc score is 2 or greater in men or 3 or greater in women.
1 B – Bleeding history or predispositiond Use of aspirin in low-risk patients is no longer recommended
in the current guidelines (January 2019, 2014). Because the
1 L – Labile INRe (TTR < 60%)
DOACs were at least noninferior and, in some cases, supe-
1 E – Elderly (age > 65 or extreme frailty) rior to warfarin in preventing stroke and systemic embolism
1a D – Drugsf or excessive alcohol consumption and were associated with lower rates of intracranial bleed-
ing in AF clinical trials, the current guidelines recommend a
Max possible score = 9
DOAC over warfarin in eligible patients for stroke prevention
Bleeding risk: 0 (low); 1 or 2 (moderate); ≥ 3 (high) in AF. Patients who have moderate to severe mitral stenosis
or a mechanical heart valve should not receive a DOAC and
a
One point for each. should instead receive warfarin. Appropriate end-organ func-
b
Dialysis, transplantation, SCr > 2.26 mg/dL, cirrhosis, tion, relevant drug-drug and drug-diet interactions, patient
bilirubin > 2 × upper limit of normal; AST/ALT/ALP > 3 ×
adherence, and accessibility should all be assessed when
upper limit of normal.
initiating OAC treatment for AF, with reassessment at peri-
c
Hemorrhagic stroke would also score 1 point under the “B”
criterion. odic intervals throughout therapy. Table 4 summarizes guide-
d
Previous major hemorrhage, anemia, or severe line recommendations for risk-based antithrombotic therapy.
thrombocytopenia. Table 5 summarizes oral anticoagulation used for stroke pre-
e
Only relevant if patient is receiving warfarin. vention in AF.
f
Concomitant use of antiplatelet or NSAID.
SBP = systolic blood pressure; TTR = time in therapeutic Symptom Management
range.
Information from: Hindricks G, Potpara T, Dagres N, et al.
For newly diagnosed AF, in concert with stroke risk assess-
2020 ESC guidelines for the diagnosis and management of ment and choice of anticoagulant, a decision should be
atrial fibrillation developed in collaboration with the made regarding whether a rate- or rhythm-control strategy
European Association for Cardio-Thoracic Surgery will be pursued. A rate-control strategy usually involves
(EACTS). Eur Heart J 2021;42:373-498.
the use  of pharmacologic agents that slow conduction

PSAP 2022 Book 1 • Cardiology 126 Management of Atrial Fibrillation


Table 4. Summary of Recommendations for Risk-Based Antithrombotic Therapy

COR LOE Recommendation

I C AC therapy should be based on shared decision-making, discussion of risk of stroke and bleeding, and
patient’s values and preferences

I B AC therapy should be selected on the basis of thromboembolism risk

I B CHA 2DS2-VASc score is recommended to assess stroke risk (exception: patients with moderate to severe
mitral stenosis or a mechanical heart valve)

IIa B For patients with CHA 2DS2-VASc score of 0 in men or 1 in women, it is reasonable to omit AC therapy

IIb C-LD For patients with CHA 2DS2-VASc score of 1 in men or 2 in women, it is reasonable to consider AC therapy

I A For patients with CHA 2DS2-VASc score ≥ 2 in men or ≥ 3 in women, AC therapy is recommended. Options
include:
• Warfarin (LOE: A)
• Dabigatran (LOE: B)
• Rivaroxaban (LOE: B)
• Apixaban (LOE: B)
• Edoxaban (LOE: B-R)

I A DOACs are recommended over warfarin in DOAC-eligiblea patients with AF

I C DOACs are recommended in DOAC-eligiblea patients if unable to maintain therapeutic INR on warfarin

I B For patients with AF who have mechanical heart valves, warfarin is recommended. Target INR intensity
should be based on type and location of valvular prosthesis

I A With warfarin, determine INR at least weekly during initiation, and monthly when stable

I B-NR With DOACs, renal and hepatic function should be evaluated before initiation and at least annually

I C Need for and choice of AC therapy should be reevaluated periodically to reassess stroke and bleeding risks

I C For patients with atrial flutter, AC therapy is recommended according to the same risk profile as used for AF

IIb B-R For patients with CHA 2DS2-VASc score ≥ 2 in men or ≥ 3 in women and who have renal dysfunction (as
defined in the text that follows), treatment with reduced doses of DOACs can be considered:
• Apixaban (SCr ≥ 1.5 mg/dL)
• Dabigatran (CrCl 15–30 mL/min)
• Rivaroxaban (CrCl ≤ 50 mL/min)
• Edoxaban (CrCl 15–50 mL/min)

IIb B-NR For patients with CHA 2DS2-VASc score ≥ 2 in men or ≥ 3 in women and who have end-stage CKD (CrCl <
15 mL/min) or are receiving dialysis, it may be reasonable to prescribe warfarin (INR 2–3) or apixaban

III: No C-EO In patients with AF and end-stage CKD or receiving dialysis, neither dabigatran, rivaroxaban, nor edoxaban
benefit is recommended because of lack of evidence that benefit exceeds risk

III: Harm B-R Dabigatran should not be used in patients with AF and a mechanical heart valve

a
Exclusion criteria for DOAC: moderate to severe mitral stenosis or a mechanical heart valve.
AC = anticoagulation; CKD = chronic kidney disease; COR = classification of recommendation; DOAC = direct oral anticoagulant;
LOE = level of evidence.
Information from: January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/ACC/HRS guideline
for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association
Task Force on Clinical Practice Guidelines and the Heart Rhythm Society in Collaboration with the Society of Thoracic Surgeons
[published correction appears in Circulation 2019;140:e285]. Circulation 2019;140:e125-e151; January CT, Wann LS, Alpert JS, et al.
2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of
Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society [published correction
appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

PSAP 2022 Book 1 • Cardiology 127 Management of Atrial Fibrillation


Table 5. Oral Anticoagulants Used in Stroke Prevention for AF

Characteristic Warfarin Dabigatran Rivaroxaban Apixaban Edoxaban

MOA Vitamin K antagonist Direct thrombin Factor Xa inhibition Factor Xa inhibition Factor Xa
inhibitor inhibition
Standard 5 mg daily (avoid loading 150 mg BID 20 mg daily with 5 mg BID 60 mg daily
dosing in AF doses), adjust to evening meal
achieve INR 2–3
Renal Consider starting doses 75 mg BID 15 mg daily 2.5 mg BID 30 mg daily
impairment of ≤ 2.5 mg daily if: (CrCl 15–30 (CrCl 15–50 mL/ If at least two of three (CrCl 15–50 mL/
dosing for AF — Age ≥ 65 mL/min) min) criteria: min)a
— Weight ≤ 70 kg — Age ≥ 80
— Poor nutritional status — Weight ≤ 60 kg
— Significant hepatic — SCr ≥ 1.5 mg/dL
Hepatic disease No adjustment Child-Pugh class B: Child-Pugh class B: Child-Pugh class
impairment — bleeding risk Avoid or use with Avoid or use with B: Avoid or use
dosing for AF — Known warfarin caution caution with caution
sensitivity Child-Pugh class C: Child-Pugh class C: Child-Pugh class
— Decompensated HF Avoid use Avoid use C: Avoid use
Time to peak 5–7 days 1–3 hr 2–4 hr 1–2 hr 1–2 hr
Half-life (hr) ~40 8–15 7–11 12 10–14
Excretion Hepatic, primarily 80% renal 66% renal (one-half 25% renal, 75% fecal 50% renal; also
through CYP2C9 as inactive form) bile, feces
Eliminated or CYP2C9, CYP1A2, P-gp P-gp P-gp P-gp
metabolized by CYP3A4, CYP2C19 CYP3A4 CYP3A4
P-gp and/or Consider higher starting Avoid use Avoid use Avoid use Avoid use
strong CYP3A4 dose, monitor INR
INDUCERSb closely
P-gp N/A If CrCl < 50 mL/ N/A N/A 30 mg daily
INHIBITORSc min, avoid use
or reduce dose
Dual P-gp and Consider lower starting N/A — Avoid use — Decrease dose by 50% N/A
strong CYP3A4 dose, monitor INR — No dose change — Avoid use if already
INHIBITORSd closely needed with on 2.5 mg BID
concomitant — No dose change
clarithromycin needed with
concomitant
clarithromycin
Dual P-gp Monitor INR closely N/A If CrCl < 80 mL/min, Use with caution N/A
and moderate avoid use unless
CYP3A4 benefit justifies
INHIBITORSe potential risk
Reversal Vitamin K, PCC, or FFP Idarucizumab Andexanet alfa Andexanet alfa Andexanet alfag
strategy PCC f PCC f PCC f

a
Avoid use in patients with CrCl > 95 mL/min.
b
For example, barbiturates, carbamazepine, dexamethasone, phenytoin, rifampin, St. John’s wort.
c
For example, amiodarone, carvedilol, diltiazem, dronedarone, macrolides, oral itraconazole or ketoconazole, quinidine, verapamil.
d
For example, clarithromycin, oral itraconazole or ketoconazole, cobicistat, indinavir, ritonavir, saquinavir, telaprevir.
e
For example, cyclosporine, diltiazem, dronedarone, erythromycin, verapamil.
f
Non–FDA-approved indication for PCC, but is recommended by guidelines as an option for reversal when andexanet alfa is unavailable.
g
Non–FDA-approved indication for andexanet alfa, but is recommended by guidelines as an option for anticoagulation reversal.
BID = twice daily; FFP = fresh frozen plasma; MOA = mechanism of action; N/A = not applicable; PCC = prothrombin complex
concentrate; P-gp = P-glycoprotein.
Information from: Lexi-Drugs. Lexicomp. Wolters Kluwer Health.

PSAP 2022 Book 1 • Cardiology 128 Management of Atrial Fibrillation


across the  atrioventricular (AV) node, slowing the ventricu- guideline recommendations for ventricular rate control.
lar response. A rhythm-control strategy may use electrical or Intensity of initial rate-control therapy depends on the clin-
pharmacologic cardioversion, percutaneous catheter abla- ical scenario, where intravenous medications are used ini-
tion, or surgical ablation. tially for symptomatic patients with a rapid ventricular
Historically, the choice between rate and rhythm control response (e.g., greater than 120 beats/minute) and oral
has largely been determined on the basis of patient-specific medications can be used upfront for those with few to no
factors. Despite several studies, differences in rates of seri- symptoms. Once initial rate control is achieved with an intra-
ous morbidity or mortality have not been shown between the venous agent, patients can be changed to oral rate-control
two approaches at the time of current guideline publication therapy.
(Hindricks 2021; January 2019, 2014). In addition, although it Choice of initial rate-control agent also depends on the
might seem intuitive that patients would feel better if main- presence of patient comorbidities, as outlined in Figure 3,
tained in normal sinus rhythm than in AF, this is not always and adverse effects. In general, β-blockers or non-dihydropy-
true. In some instances, however, rhythm control can be ridine calcium channel blockers are considered first-line ther-
considered a preferred strategy. Symptomatic patients or apy for rate control, followed by amiodarone and digoxin as
those with presumed tachycardia-induced cardiomyopathy second line. β-Blockers are the most commonly used agents,
should be considered for rhythm control in order to prevent largely because of better rate control; however, calcium chan-
the irreversible structural and electrical remodeling that can nel blockers can provide reasonable rate control and better
occur with longstanding persistent AF. For similar reasons, a improve AF-related symptoms than β-blockers (Hindricks
rhythm-control approach may also be preferable in younger 2021). Combination therapy of β-blockers and calcium chan-
patients (younger than 65) because rate control alone is nel blockers can be considered in patients for whom ade-
likely to result in progression to longstanding persistent AF quate ventricular rate control cannot be achieved with a
(Ponamgi 2021). In asymptomatic patients with longstanding single agent; however, both heart rate and blood pressure
persistent AF or in those for whom a rhythm-control strategy should be monitored closely. Both β-blockers and calcium
has failed or who cannot tolerate a rhythm-control strategy, a channel blockers should be avoided in the setting of decom-
rate-control approach is reasonable. pensated HF and in hemodynamically unstable patients (Hin-
Recently published, the EAST-AFNET 4 trial provides con- dricks 2021; January 2014).
temporary evidence in support of an early rhythm-control Digoxin is considered a second-line agent for rate control
strategy. The authors randomized 2789 patients with early for several reasons. Digoxin has a slow onset of action (greater
AF (defined as AF diagnosed within 12 months before enroll- than 1 hour with intravenous administration), and effects do
ment, with a median time since diagnosis of 36 days) and at not peak until 6 hours after initial administration, making it
high risk of CV complications to either early rhythm control ineffective for acute rate control. In addition, digoxin is inef-
with antiarrhythmic drugs (AADs) or ablation or usual care, fective in patients with increased sympathetic tone, making it
which limited rhythm control to patients unable to tolerate ineffective in controlling ventricular rate during exercise (Hin-
rate control. The trial was terminated early for efficacy after dricks 2021; January 2014). Observational studies have also
a median of 5.1 years of follow-up because patients in the associated long-term digoxin therapy with increased mortal-
rhythm-control group had a significantly lower incidence of ity in patients with AF; however, these findings may be sub-
the composite clinical end point, including CV death, stroke, ject to selection bias because digoxin has historically been
or hospital admission for HF or acute coronary syndrome prescribed most commonly to patients with more severe
(ACS), than patients in the rate-control group (3.9 vs. 5.0 disease. Lower digoxin doses with serum concentrations of
per 100 person-years; HR 0.79; 95% CI, 0.66–0.94; p=0.005) 0.9  ng/mL or less have been associated with a better prog-
(Kirchhof 2020). nosis in patients with AF (Hindricks 2021). Recently, in the
Figure 2 outlines a general approach to the management RATE-AF trial, 160 patients with permanent AF and symp-
of new-onset AF. Unstable patients with signs of hemody- toms of HF were randomized to either digoxin or bisoprolol.
namic instability, severe symptoms, or decompensated HF Although the primary end point of patient-reported quality
should be emergently treated with electrical cardioversion, of life did not differ, digoxin showed significant improvement
followed by therapeutic anticoagulation for at least 4 weeks. in other secondary quality-of-life measures at 12 months,
In stable patients, a strategy of rate or rhythm control should greater reductions in New York Heart Association (NYHA)
be determined (see Figure 2). class and N-terminal pro–brain natriuretic peptide, and fewer
adverse events than bisoprolol (Kotecha 2020). Of note, only
Rate Control 19% of patients had an LVEF less than 50% at baseline in the
Slowing the ventricular response to AF is important for RATE-AF trial. Although the clinical significance of this trial
many reasons – namely, reducing symptoms and increas- has not yet been determined, it provides much-needed data in
ing quality of life together with reducing the potential for the area of permanent AF treatment and assessed clinically
tachycardia-induced cardiomyopathies. Table 6 summarizes important outcomes.

PSAP 2022 Book 1 • Cardiology 129 Management of Atrial Fibrillation


New-Onset AF

UNSTABLE STABLE

Emergency DCCV Rhythm Control Rate Control


Followed by ≥ 4 wk AC,
with long-term AC according
to stroke/bleeding risk
Duration < 48 hr Duration ≥ 48 hr or unknown β-Blocker
CCB
Digoxin
Amiodaronee
Initiate UFH, Delayed cardioversion: (see Figure 3)
LMWH, or DOACa • AC × 3 wk, then DCCV WITH
• AC ≥ 4 wk post DCCV
AC according
OR to stroke risk
Electrical Pharmacologic
cardioversion: Early cardioversion:
cardioversion
• Initiate UFH, LMWH, or
(DCCV)b Amiodarone DOAC
Dofetilide • TEE to r/o LA thrombus
Ibutilidec – If no LA thrombus → try
Flecainided cardioversion
Propafenoned – If LA thrombus →
continue AC ≥ 4 wk,
then repeat TEE

Long-term AC according to stroke/bleeding risk profile

Figure 2. Approach to management of new-onset atrial fibrillation.


a
Can consider no anticoagulation in patients with a CHA 2DS2-VASc score of 0 in men or 1 in women.
b
Patients should not be sedated for synchronized DCCV if they have eaten a meal within 12 hr because of the risk of aspiration.
c
Ibutilide can be administered to patients with left ventricular ejection fraction (LVEF) 30%–40% but should be avoided in patients
with LVEF < 30% because of the risk of ventricular proarrhythmia.
d
Avoid use of flecainide or propafenone in structural heart disease (coronary artery disease, LVEF < 40%).
e
When used for rate control, amiodarone may result in pharmacologic cardioversion; consider LA thrombus r/o, and ensure
therapeutic anticoagulation, if possible, before amiodarone initiation.
CCB = calcium channel blocker; DCCV = direct current cardioversion; DOAC = direct oral anticoagulant; HF = heart failure; LA = left
atrium; LMWH = low-molecular-weight heparin; r/o = rule out; TEE = transesophageal echocardiogram; UFH = unfractionated heparin.
Information from: January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/ACC/HRS guideline
for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association
Task Force on Clinical Practice Guidelines and the Heart Rhythm Society in Collaboration with the Society of Thoracic Surgeons
[published correction appears in Circulation 2019;140:e285]. Circulation 2019;140:e125-e151; January CT, Wann LS, Alpert JS, et al.
2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of
Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society [published correction
appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

Amiodarone is a last-line option when ventricular rate can- In addition, although initial use may be intended for rate con-
not be controlled by other therapies or combination therapies, trol, amiodarone may result in unintentional pharmacologic
or in critically ill patients with severely impaired LV systolic cardioversion. Careful consideration should be given to eval-
function. Amiodarone has several potential toxicities and uating the duration of AF, presence of LA thrombus, and use
drug interactions together with a high monitoring burden that of therapeutic anticoagulation when initiating amiodarone
limit its use long term in a ventricular rate-control strategy. therapy. Table 7 summarizes the dosing, monitoring, and

PSAP 2022 Book 1 • Cardiology 130 Management of Atrial Fibrillation


Table 6. Summary of Recommendations for Ventricular Rate Control

COR LOE Recommendation

I B Control ventricular rate using a β-blocker or non-DHP CCB for paroxysmal, persistent, or permanent AF

I B IV β-blockers or non-DHP CCBs are recommended to slow ventricular rate in the acute setting in patients
without preexcitation. In hemodynamically unstable patients, electrical cardioversion is indicated

1 C For AF, assess HR control during exertion and adjust pharmacologic treatment as necessary

IIa B An HR control (resting HR < 80 beats/min) strategy is reasonable for symptomatic management of AF

IIa B IV amiodarone can be useful for rate control in critically ill patients without preexcitation

IIa B AV nodal ablation with permanent ventricular pacing is reasonable when pharmacologic therapy is
inadequate and rhythm control is not achievable

IIb B A lenient rate-control strategy (resting HR < 110 beats/min) may be reasonable when patients remain
asymptomatic and LV systolic function is preserved

IIb C Oral amiodarone may be useful for ventricular rate control when other measures are unsuccessful or
contraindicated

III: Harm C AV nodal ablation should not be performed without prior attempts to achieve rate control with medications

III: Harm C Non-DHP CCBs should not be used in decompensated HF

III: Harm B With preexcitation and AF, digoxin, non-DHP CCBs, or amiodarone should not be administered

III: Harm B Dronedarone should not be used to control ventricular rate with permanent AF

AF = atrial fibrillation; AV = atrioventricular; CCB = calcium channel blocker; DDI = drug-drug interaction; DHP = dihydropyridine; HF =
heart failure; HR = heart rate; IV = intravenous(ly).
Information from: January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial
fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the
Heart Rhythm Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

clinical considerations of various pharmacologic ventricular of AF, AF precipitated by an acute illness, younger age, and
rate-control therapies. patient preference (Hindricks 2021; January 2014).
Ultimately, as last line, a nonpharmacologic strategy of AV Electrical cardioversion is performed using direct current
nodal ablation can be considered in patients for whom prior energy in the form of an electrical shock, delivered through
attempts with rate-control therapy have failed and in whom electrodes placed on the patient’s chest. Energy delivery is
synchronized with the QRS complex to avoid delivery during
rhythm control cannot be achieved. However, because AV
the ventricular refractory period (the ST segment), which
nodal ablation results in pacemaker dependency, patients
can potentiate ventricular arrhythmias. Sedation is typically
should be monitored closely for appropriate device follow-up
given for patient comfort; however, in an emergency, seda-
and maintenance.
tion may not be possible. In unstable patients, electrical car-
dioversion is the preferred rhythm control because it is more
Rhythm Control
effective than pharmacologic cardioversion and results in
A rhythm-control strategy tries to restore and maintain immediate restoration of sinus rhythm. In stable patients,
normal sinus rhythm using a combination of treatment either electrical or pharmacologic cardioversion can be
approaches, including electrical cardioversion, AADs, cath- considered. Although pharmacologic cardioversion is less
eter ablation, or surgical ablation, together with adequate effective, it does not require the use of sedation. However,
rate control and anticoagulant therapy. Several consider- pretreatment with AADs may improve the efficacy of electri-
ations favor pursuit of a rhythm-control strategy, but per- cal cardioversion.
sistent AF symptoms and the goal of improving quality of life Management of anticoagulation around the time of cardio-
are the most common driving factors. Other situations that version is important to reduce thromboembolic risk. Longer
may favor rhythm control include difficulty maintaining rate AF durations (48 hours or more) are more likely to result in the
control, tachycardia-mediated cardiomyopathy, first episode formation of LA thrombi, which can migrate in response to

PSAP 2022 Book 1 • Cardiology 131 Management of Atrial Fibrillation


Atrial Fibrillation

No other CV HTN or LV dysfunction


HFpEF COPD
disease or HF

β-Blocker β-Blocker β-Blocker


β-Blockera
Diltiazem Diltiazem Diltiazem
Digoxinb
Verapamil Verapamil Verapamil

Amiodaronec

Figure 3. Approach to selecting therapy for ventricular rate control. Drugs are listed alphabetically.
a
β-Blockers should be instituted after stabilization of patients with decompensated HF. Choice of β-blocker (e.g., cardioselective)
depends on the patient’s clinical condition.
b
Digoxin is not usually first-line therapy. It can be combined with a β-blocker and/or a non-DHP CCB when ventricular rate control is
insufficient and may be useful in patients with HF.
c
Partly because of concern over its adverse effect profile, use of amiodarone for chronic rate control of ventricular rate should be
reserved for patients who do not respond to or are intolerant of β-blockers or non-DHP CCBs.
CCB = calcium channel blocker; COPD = chronic obstructive pulmonary disease; CV = cardiovascular; DHP = dihydropyridine;
HFpEF = heart failure with preserved ejection fraction; LV = left ventricular.
Information from: January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial
fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the
Heart Rhythm Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

cardioversion and restoration of organized atrial contraction. Similarly, in the X-VeRT trial, patients with new-onset AF were
De novo atrial thrombi may also form after electrical cardio- administered the first dose of a rivaroxaban 20-mg/day reg-
version as a result of atrial stunning and depressed mechan- imen at least 4 hours before cardioversion (Cappato 2014).
ical function. Thus, confirmation of both the duration of AF For patients in AF for 48 hours or more or if the duration
and the presence of LA thrombi is key in determining the most of AF is unknown, anticoagulation with either warfarin or a
appropriate cardioversion strategy. DOAC should be administered for 3 weeks before cardiover-
Table 8 summarizes guideline recommendations for elec- sion, given the increased risk of thrombus development. Per-
trical and pharmacologic cardioversion of AF together with
forming a transesophageal echocardiogram (TEE) to exclude
maintenance of sinus rhythm. In patients at low throm-
the presence of LA thrombus is an alternative to 3 weeks of
boembolic risk (CHA 2DS2-VASc of 0 in men or 1 in women),
anticoagulation before cardioversion; if no thrombus is seen,
administration of intravenous unfractionated heparin (UFH),
cardioversion can be performed as long as therapeutic anti-
low-molecular-weight heparin (LMWH), a DOAC, or no antico-
coagulation is achieved at the time of cardioversion. If a
agulation can be considered before cardioversion (electrical
or pharmacologic), without the need for post-cardioversion thrombus is identified, cardioversion should be postponed,
anticoagulation. For patients in AF for less than 48 hours with anticoagulation continued for at least 3 weeks before
and a CHA 2DS2-VASc of 2 or greater in men or 3 or greater retrying cardioversion. A TEE may be repeated to ensure
in women, anticoagulation with intravenous UFH, LMWH, or thrombus resolution before retrying cardioversion. For unsta-
a DOAC should be initiated as soon as possible before car- ble patients in AF requiring emergency cardioversion, antico-
dioversion, with significant consideration for achieving ther- agulation should be initiated as soon as possible but should
apeutic serum drug concentrations before the procedure. For not delay interventions to stabilize the patient. After either
example, in the EMANATE trial, patients with new-onset AF electrical or pharmacologic cardioversion, anticoagulation
could undergo cardioversion no sooner than 2 hours after should be continued for at least 4 weeks unless contraindi-
receiving one loading dose of apixaban 10 mg or five consec- cated, and decisions regarding long-term anticoagulation for
utive doses of apixaban 5 mg twice daily (Ezekowitz 2018). all patients with AF should be based on the thromboembolic

PSAP 2022 Book 1 • Cardiology 132 Management of Atrial Fibrillation


Table 7. Drugs Used for Ventricular Rate Control of AF

Maintenance Dose
Drug Loading Dose (IV) (PO) Adverse Effects Notes

β-Blockersa

Metoprolol 2.5- to 5-mg IV bolus over 2 min, 25–100 mg BID AV block Use β1-selective
tartrate up to three doses Bradycardia blockers in asthma,
HF exacerbation COPD
Metoprolol N/A 50–400 mg daily
Hypotension
succinate CI in ADHF and
history of severe
Esmolol 500-mcg IV bolus over 1 min, N/A
bronchospasm
then 50–300 mcg/kg/min

Propranolol 1-mg IV bolus over 1 min; up to 10–30 mg TID or QID Inhibit lidocaine
three doses at 2-min intervals clearance

Non-DHP Calcium Channel Blockers

Diltiazem 0.25-mg/kg IV bolus over 2 min, 120–360 mg once daily AV block CI in HFrEF
then 5–15 mg/hr (ER) Bradycardia
Adjust doses in
HF exacerbation
Verapamil 0.075- to 0.15-mg/kg IV bolus 180–480 mg once hepatic and renal
Hypotension
over 2 min; then 0.005 daily (ER) impairment
Constipationb
mg/kg/min (rarely used)
DDIs

Others

Digoxin 0.25- to 0.5-mg IV bolus, repeat 0.125–0.25 mg once Anorexia Not dialyzable,
with doses of 0.25 mg every 6 hr daily N/A adjust doses in
up to a max of 1.5 mg over 24 hr Ventricular arrhythmias renal impairment

Amiodarone 300 mg IV over 1 hr, then 10–50 100–200 mg once daily AV block May exacerbate
mg/hr over 24 hr (0.5–1 mg/min) Bradycardia thyroid disease
Hypotension (IV)
DDIs
Hypo/hyperthyroidism
Hepatotoxicity
Pulmonary fibrosis
Photosensitivity
Blue-gray skin
discoloration
Corneal microdeposits

a
Not an all-inclusive list.
b
Specific to oral verapamil only.
ADHF = acute decompensated heart failure; AV = atrioventricular; CI = contraindicated; DDI = drug-drug interaction; ER = extended
release; HFrEF = heart failure with reduced ejection fraction; IV = intravenous(ly); PO = oral(ly); QID = four times daily; TID = three
times daily.
Information from: January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial
fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the
Heart Rhythm Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76;
Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of atrial fibrillation developed in
collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J 2021;42:373-498.

and bleeding risk profile using the CHA 2DS2-VASc and HAS- after onset of AF. Table 9 lists selected AADs used in phar-
BLED scores for risk assessment (January 2019, 2014). macologic cardioversion of AF, and Table 10 outlines AADs
Antiarrhythmic drugs can be used for pharmacologic car- used to maintain sinus rhythm. Selection of AAD therapy is
dioversion or as adjunctive therapy to increase the efficacy largely driven by concerns regarding drug safety rather than
of electrical cardioversion. Efficacy of pharmacologic car- drug efficacy, and selection of AADs is largely based on type
dioversion varies but is highest when initiated within 7 days of severity of associated comorbidities. Figure 4 outlines a

PSAP 2022 Book 1 • Cardiology 133 Management of Atrial Fibrillation


Table 8. Summary of Recommendations for Electrical and Pharmacologic Cardioversion of AF and Atrial Flutter and
Maintenance of Sinus Rhythm

COR LOE Recommendation

Prevention of Thromboembolism

I B-R With AF or atrial flutter for ≥ 48 hr or unknown duration, anticoagulation with warfarin (INR 2–3) or a DOAC is
recommended for at least 3 wk before and 4 wk after cardioversion, regardless of the CHA 2DS2-VASc score
or the method (electrical or pharmacologic) used to restore sinus rhythm

I C With AF or atrial flutter > 48 hr or unknown duration requiring immediate cardioversion for hemodynamic
instability, anticoagulation should be initiated as soon as possible and continued for at least 4 wk unless CI

IIa B-NR With AF or atrial flutter < 48 hr and a CHA 2DS2-VASc score ≥ 2 in men and ≥ 3 in women, administration of
IV UFH, LMWH, or a DOAC is reasonable as soon as possible before cardioversion, followed by long-term
anticoagulation

I C-EO After cardioversion of AF of any duration, the decision about long-term anticoagulation should be based on
the thromboembolic and bleeding risk profile

IIa B With AF or atrial flutter ≥ 48 hr or unknown duration and no anticoagulation for the preceding 3 wk, it is
reasonable to perform TEE before cardioversion and then cardiovert if no LA thrombus is identified (including
in the LAA) if anticoagulation is achieved before TEE and maintained after cardioversion for at least 4 wk

IIb B-NR With AF or atrial flutter < 48 hr and a CHA 2DS2-VASc score of 0 in men or 1 in women, administration of IV
UFH, LMWH, a DOAC, or no anticoagulation can be considered before cardioversion, without the need for
post-cardioversion anticoagulation

Direct Current Cardioversion

I B Cardioversion is recommended for AF or atrial flutter to restore sinus rhythm. If successful, cardioversion
attempts can be repeated

I C Cardioversion is recommended for AF or atrial flutter with RVR that does not respond to pharmacologic
therapies

I C Cardioversion is recommended for AF or atrial flutter and preexcitation with hemodynamic instability

IIa C It is reasonable to repeat cardioversion in persistent AF when sinus rhythm can be maintained for a clinically
meaningful time between procedures

Pharmacologic Cardioversion

I A Flecainide, dofetilide, propafenone, and IV ibutilide are useful for cardioversion for AF or atrial flutter if
contraindications to the selected drug are absent

IIa A Amiodarone is reasonable for pharmacologic conversion of AF

IIa B Propafenone or flecainide (“pill-in-the-pocket”) to terminate AF out of hospital is reasonable once observed
to be safe in a monitored setting

III: Harm B Dofetilide should not be initiated out of hospital

Maintenance of Sinus Rhythm

I C Before initiating AAD therapy, treatment of precipitating or reversible causes of AF is recommended

I A The following AADs are recommended in patients with AF to maintain NSR: amiodarone, dofetilide,
dronedarone, flecainide, propafenone, and sotalol

I C Risks of the AAD, including proarrhythmia, should be considered before initiating therapy

I C Because of potential toxicities, amiodarone should only be used after consideration of risks and when other
agents have failed or are CI

IIa C A rhythm-control strategy with pharmacologic therapy can be useful in patients with AF for the treatment of
tachycardia-induced cardiomyopathy

(continued)

PSAP 2022 Book 1 • Cardiology 134 Management of Atrial Fibrillation


Table 8. Summary of Recommendations for Electrical and Pharmacologic Cardioversion of AF and Atrial Flutter and
Maintenance of Sinus Rhythm  (continued)

COR LOE Recommendation

IIb C It may be reasonable to continue current AAD therapy in the setting of infrequent, well-tolerated recurrences
of AF when the drug has reduced the frequency of symptoms of AF

III: Harm B AADs for rhythm control should not be continued when AF becomes permanent, including dronedarone

III: Harm B Dronedarone should not be used for treatment of AF in patients with NYHA class III or IV HF or patients who
have had an episode of decompensated HF in the past 4 wk

AAD = antiarrhythmic drug; AF = atrial fibrillation; CI = contraindicated; DOAC = direct oral anticoagulant; HF = heart failure; LAA = left
atrial appendage; NSR = normal sinus rhythm; RVR = rapid ventricular response.
Information from: January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/ACC/HRS guideline
for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association
Task Force on Clinical Practice Guidelines and the Heart Rhythm Society in Collaboration with the Society of Thoracic Surgeons
[published correction appears in Circulation 2019;140:e285]. Circulation 2019;140:e125-e151 January CT, Wann LS, Alpert JS, et al.
2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of
Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society [published correction
appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

Table 9. Drugs Used for Pharmacologic Conversion of AF

Drug Loading Dose Maintenance Dose Adverse Effects Notes

Amiodarone 600–800 mg PO daily in 200 mg PO daily Phlebitis (IV) Administer through a large
divided doses to a total Hypotension peripheral vein or central
load of up to 10 ga Bradycardia vein to minimize phlebitis
AV block risk; avoid infusions > 24 hr
150 mg IV over 10 min, then
QTc interval
1 mg/min × 6 hr, then 0.5
TdP (rare)
mg/min × 18 hr or change
GI upset
to PO dosing

Dofetilide CrCl (mL/min): Initial dosing: Measure QTc 2–3 hr after QTc interval Patients must be
> 60: 500 mcg BID first dose: If QTc increases TdP (3%–4%) hospitalized for at least
40–60: 250 mcg BID to > 15% above baseline or 3 days (≥ 5 doses) during
20–39: 125 mcg BID > 500 ms (> 550 ms with initiation of therapy
< 20: Do not use conduction abnormalities),
See Table 10 for relevant
reduce dose by 50%
Do not initiate if baseline DDIs
QTc > 440 ms (or > Monitor QTc 2–3 hr after
500 ms in presence of each subsequent in-hospital
ventricular conductional dose 2-5. If QTc > 500 ms
abnormalities) (> 550 ms with conduction
abnormalities) at any time,
discontinue dofetilide

Ibutilide 1 mg IV over 10 min; may None QTc interval Consider pretreatment with
repeat × 1 if needed TdP IV magnesium to lower TdP
(if weight < 60 kg, use risk
0.01 mg/kg)
CI in prolonged QTc
(> 440 ms), severe LVH, or
EF < 30%

(continued)

PSAP 2022 Book 1 • Cardiology 135 Management of Atrial Fibrillation


Table 9. Drugs Used for Pharmacologic Conversion of AF  (continued)

Drug Loading Dose Maintenance Dose Adverse Effects Notes

Flecainide 200–300 mg single PO dose None (for conversion to sinus Blurred vision CI in CAD and structural
(pill-in-the-pocket)b rhythm) Dizziness heart disease
HF exacerbation
QRS
Ventricular
arrhythmias
Atrial flutter with
1:1 AV conduction

Propafenone 450–600 mg single PO dose None (for conversion to sinus AV block CI in CAD and structural
(pill-in-the-pocket)b rhythm) Bradycardia heart disease
HF exacerbation
QRS
Ventricular
arrhythmias
Atrial flutter with
1:1 AV conduction

a
Many dosing schemes exist for amiodarone.
b
Recommended to administer together with a β-blocker or non-DHP CCB given ≥ 30 min before administering the AAD to prevent
RVR because of 1:1 AV conduction during atrial flutter.
AV = atrioventricular; CI = contraindicated; DDI = drug-drug interaction; LVH = left ventricular hypertrophy; ms = millisecond(s);
TdP = torsades de pointes.
Information from: January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial
fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the
Heart Rhythm Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76; Tisdale
JE. Arrhythmias. In: Updates in Therapeutics®: Cardiology Pharmacy Preparatory Review Course. American College of Clinical
Pharmacy, 2018:447-524.

Table 10. Drugs Used to Maintain Sinus Rhythm in Patients with AF

Recommended
Drug Dose Adverse Effects Monitoring Major DDIs

Amiodarone Loading dose: AV block LFTs – baseline and Inhibits CYP1A2, CYP2C9,
400–600 mg/day PO Bradycardia every 6 mo CYP2D6, and CYP3A4:
in two or three divided Hepatotoxicity Increases warfarin
TFTs – baseline and
doses for 2–4 wka Hyperthyroidism and (some) statin
every 6 mo
Hypothyroidism concentrations
Maintenance dose:
Pulmonary fibrosis PFTs – baseline, for Inhibits P-gp: Increases
100–200 mg PO once
Photosensitivity unexplained dyspnea digoxin concentrations
daily
Blue-gray skin
SCr, electrolytes –
discoloration
baseline and as indicated
Corneal microdeposits
QTc interval Chest radiography –
TdP (rare) baseline and annually
GI upset
ECG – baseline and
annually

Ophthalmologic
examination – at
baseline and for any
symptoms

(continued)

PSAP 2022 Book 1 • Cardiology 136 Management of Atrial Fibrillation


Table 10. Drugs Used to Maintain Sinus Rhythm in Patients with AF  (continued)
Recommended
Drug Dose Adverse Effects Monitoring Major DDIs

Dofetilide CrCl (mL/min): Dosing: QTc interval Continuous ECG Renal tubular secretion
> 60: 500 mcg PO BID TdP monitoring during first and concentrations
40–60: 250 mcg PO BID 3 days of dosing while increased by cimetidine,
20–39: 125 mcg PO BID hospitalized HCTZ, ketoconazole,
< 20: Do not use itraconazole, megestrol,
ECG every 3–6 mo
prochlorperazine,
SCr every 3–6 mo trimethoprim,
dolutegravir, Biktarvy,
and verapamil
(concomitant use CI)
Dronedarone 400 mg PO BID Bradycardia LFTs – baseline and Inhibits CYP3A: Increases
Diarrhea every 6 mo concentrations of (some)
Hepatotoxicity ECG every 3 mo statins, verapamil, and
Nausea SCr every 3–6 mo diltiazem
Pulmonary fibrosis
Inhibits CYP2D6:
Worsening HF
Increases concentrations
of β-blockers, other
CYP2D6 substrates
Inhibits P-gp: Increases
dabigatran and digoxin
concentrations
Sotalol CrCl (mL/min): Initial Bradycardia Continuous ECG None; however, sotalol
dosing: QTc interval monitoring during the may have an additive
> 60: 80 mg PO BID TdP first 3 days of dosing effect with other drugs
40–60: 80 mg PO daily while hospitalized that prolong the QT
< 40: Do not use interval or cause sinus
ECG every 3–6 mo
bradycardia or AV block
Maintenance dosing:
SCr every 3–6 mo
If 80-mg doses are
tolerated and QTc
remains < 500 ms after
3 days, patient can be
discharged. Alternatively,
dose can be increased
to 120 mg once or twice
daily as appropriate
during hospitalization
and patient followed for
3 days on this dose
Propafenone Immediate release: AV block ECG as needed, at least Inhibits P-gp: Increases
150–300 mg PO every 8 hr Bradycardia every 6 mo digoxin concentrations
HF exacerbation
Extended release: Inhibits CYP2C9:
QRS
225–425 mg PO every Increases warfarin
Ventricular arrhythmias
12 hr concentrations
Flecainide 50–200 mg PO every 12 hr Blurred vision ECG as needed, at least Inhibitors of CYP2D6
Dizziness every 6 mo may increase serum
HF exacerbation flecainide concentrations
QRS Amiodarone increases
Ventricular arrhythmias serum flecainide
concentrations

a
Many dosing schemes exist for amiodarone.
CI = contraindicated; HCTZ = hydrochlorothiazide; LFT = liver function test; PFT = pulmonary function test; TFT = thyroid function test.
Information from: January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial
fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the
Heart Rhythm Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76; Tisdale
JE. Arrhythmias. In: Updates in Therapeutics®: Cardiology Pharmacy Preparatory Review Course. American College of Clinical
Pharmacy, 2018:447-524.

PSAP 2022 Book 1 • Cardiology 137 Management of Atrial Fibrillation


No Structural Heart Disease Structural Heart Disease

CAD HF

Dofetilidec,d
Dronedarone
Flecainidec,e Catheter Dofetilidec,d Catheter Amiodarone
Propafenonee ablationb Dronedarone ablationb Dofetilidec,d
Sotalolc,d Sotalolc,d

Amiodarone Amiodarone

Figure 4. Approach to selecting therapy for maintenance of sinus rhythm in patients with paroxysmala and persistent
AF. Drugs are listed alphabetically.
a
Catheter ablation is only recommended as first-line therapy for patients with paroxysmal AF (class IIa recommendation).
b
Depending on patient preference when performed in experienced centers.
c
Not recommended with severe LVH (wall thickness > 1.5 cm).
d
Should be used with caution in patients at risk of torsades de pointes ventricular tachycardia.
e
Should be combined with AV nodal-blocking agents.
AV = atrioventricular; CAD = coronary artery disease; HF = heart failure.
Information from: January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial
fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the
Heart Rhythm Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

general approach to choosing an AAD, given concomitant management of OSA are recommended to reduce AF incidence,
disease states and risk factors. Because the efficacy of AAD progression, recurrence, and symptoms (Hindricks 2021).
therapy is usually modest and AF recurrences are common
(and often asymptomatic), a rhythm-control strategy should CONTEMPORARY STRATEGIES
not result in cessation of background anticoagulation, rate FOR STROKE PREVENTION
control, or treatment of underlying comorbidities. Although anticoagulant therapy can significantly reduce the
risk of stroke in patients with AF, it does not eliminate risk.
Concomitant Disease and Risk Factor
Adverse effects, including bleeding, may also be a significant
Management
limitation of long-term anticoagulant use for many patients.
Appropriate management of concomitant disease states and
Of interest, most strokes in AF occur primarily as a result of
AF risk factors complements stroke prevention and reduces
thromboembolism originating from the LA, specifically within
AF burden and symptom severity (Hindricks 2021). In the
the LAA. Lack of organized atrial contraction in AF contrib-
recently published RACE 3 trial, targeted therapy for under-
utes to blood stasis, increasing the risk of clot formation. The
lying conditions improved sinus rhythm maintenance in
area of the LA with the lowest state of blood flow, and thus
patients with persistent AF and HF (Rienstra 2018).
the highest rate of thrombus, is the LAA. Given the limitations
Specific recommendations are made by both the ACC/AHA
of anticoagulation and that the LAA is the primary source of
and the ESC guidelines regarding optimizing key risk factors
thromboembolism in AF, mechanical isolation of the LAA has
and concomitant disease states. In patients with hypertension
been an evolving strategy with the goal of reducing the risk of
or obstructive sleep apnea (OSA), opportunistic screening for
thromboembolism. Closure of the LAA can be accomplished
AF is recommended. If AF is present in the setting of hyperten-
either percutaneously or surgically using a wide variety of
sion, optimal blood pressure control is important to reduce AF
devices and techniques.
recurrence, together with stroke risk and bleeding. In overweight
patients or those with obesity, weight loss is recommended to
Percutaneous LAA Occlusion or Exclusion
reduce AF symptom burden, severity, and cumulative AF dura-
Percutaneous LAA closure devices have mainly been devel-
tion. Finally, alcohol avoidance, physical activity, and optimal
oped as nonpharmacologic alternatives to anticoagulation for

PSAP 2022 Book 1 • Cardiology 138 Management of Atrial Fibrillation


stroke prevention in AF. Percutaneous LAA closure devices Reddy 2014). A meta-analysis of these two trials showed that
may prevent stroke by occluding the LAA and preventing patients receiving the WATCHMAN device had significantly
thrombus formation. There are two general approaches to fewer hemorrhagic strokes than those receiving warfarin, but
percutaneous closure of the LAA. The first approach uses more ischemic strokes. However, when periprocedural events
devices that are inserted into the LA to occlude the LAA, were excluded, the difference in ischemic strokes between
or act as a plug. These include the WATCHMAN Left Atrial groups was not significant (Holmes 2015). Therefore, for
Appendage System (Boston Scientific) and the Amplatzer patients who are poor candidates for long-term anticoagulant
Amulet device (St. Jude Medical). These devices are inserted therapy (i.e., bleeding, drug intolerance, or adherence issues),
percutaneously through a catheter delivery system by fem- the WATCHMAN device may provide an alternative method of
oral venous access and are deployed into the LAA through stroke prevention (January 2019).
transseptal atrial puncture. During the procedure, echocardi-
ography and fluoroscopy are used to guide device placement Complications
and positioning. The WATCHMAN device is anchored within Both device- and procedure-related complications may occur
the LAA through a self-expanding nitinol cage with barbs that with percutaneous LAA closure, which can range from mild
attach to the endocardium, whereas the Amplatzer Amulet to life threatening. In general, complications involving LAA
consists of a nitinol mesh disc to seal the ostium of the LAA closure can be classified as access related, device implant
and a nitinol mesh distal lobe with hooks to anchor the device related, or antithrombotic treatment related.
within the LAA (January 2014). Access-related bleeding is typically the most common
The second approach for percutaneous LAA closure is to adverse event within interventional cardiology and may
tie off the LAA using an epicardial snare. The LARIAT Loop require transfusion or surgical intervention, depending on
Applicator (SentreHEART) is a suture delivery device origi- severity. Access-related bleeding events may include groin
nally designed to close a variety of soft tissue surgical wounds hematomas, femoral arterial pseudoaneurysms, and retro-
that has been adapted for use in AF. Unlike the WATCHMAN peritoneal bleeding. Moreover, in percutaneous LAA closure
and Amplatzer Amulet, the LARIAT is a non-implant device procedures, large delivery sheaths are used, placing patients
that combines a percutaneous epicardial and endocardial at higher risk of access-related events.
approach to ligate the LAA with a pre-tied polyester suture. Device implant–related complications vary widely with
Placement of the LARIAT requires percutaneous placement device type and user technique. In the PROTECT AF trial,
of a magnetic guide within the LAA to guide an epicardially overall device-related complications with WATCHMAN place-
placed lasso (through subxiphoid access) to tie off the LAA. ment were 8.7%, with a specific rate of pericardial effusion
Patients with unusual LAA anatomy or those who have had of 4.5%, with 3.3% requiring pericardiocentesis or surgical
prior cardiac surgery are not typically candidates for the intervention (Reddy 2014). More recent data analyses from
LARIAT procedure (January 2014). the EWOLUTION registry describe a 4.1% rate of pericar-
dial effusion with only 1.4% requiring surgical management
Indications and no associated deaths, likely because of increased user
Currently, in the United States, only the WATCHMAN device experience and mastery of implant techniques (Boersma
is FDA approved for LAA closure and stroke prevention 2016). Other, less common procedural complications include
in patients with AF. The Amplatzer Amulet device has CE periprocedural stroke/transient ischemic attack (TIA), device
Mark approval in Europe for LAA closure but is not currently migration or dislodgement, and cardiac perforation with
approved in the United States for this indication. However, possible cardiac tamponade.
the global, multicenter CATALYST trial is currently under Given the difference in implant approach with the LARIAT
way to compare the effectiveness of the Amulet device with device compared with occlusion devices, most complications
DOACs as an alternative treatment in an expanded popula- with LARIAT placement are related to subxiphoid access to
tion of patients with AF. The LARIAT device is FDA approved the epicardium and include possible cardiac perforation,
for suture placement and knot tying in surgical applications pericardial effusion or tamponade, and severe pericarditis.
where soft tissues are being approximated; however, it is not Although initial single-center data with LARIAT placement
specifically approved for LAA closure. showed a high rate of procedural success and minimal com-
Currently, both the U.S. and European guidelines state that plications, subsequent multicenter results have not been as
percutaneous LAA occlusion can be considered in patients favorable. In 2015, the FDA issued a safety communication
with AF at an increased risk of stroke who have contraindica- stating that cases of death and complications such as lacer-
tions to long-term anticoagulation (IIb, B-NR) (January 2019). ation or perforation of the heart or complete LAA detachment
This recommendation primarily stems from data comparing associated with the use of LARIAT had been reported. These
the WATCHMAN device with warfarin in patients with AF at concerns surrounding the procedural safety of the LARIAT
an increased risk of stroke in two randomized controlled have largely limited its use in the United States in patients
trials – the PROTECT AF and PREVAIL studies (Holmes 2014; with absolute contraindications to anticoagulation or those

PSAP 2022 Book 1 • Cardiology 139 Management of Atrial Fibrillation


with LAA anatomies unsuitable for endovascular closure with lifelong aspirin was evaluated. During a mean follow-up of 14.4
occlusion devices. months, the primary efficacy outcome of all-cause stroke or
systemic embolism occurred at a rate of 2.3% per year and
Antithrombotic Management ischemic stroke at a rate of 1.7% per year, which was lower
The role of antithrombotic therapy after LAA occlusion is than predicted rates for matched cohorts taking either aspirin
unclear, with current strategies varying widely and based (7.3%) or clopidogrel (5%) (Reddy 2013). In addition, data from
solely on historical studies, device type, and expert opinion. EWOLUTION registry noted that of the 1021 patients receiving
Given the results of the PROTECT AF and PREVAIL trials, the a WATCHMAN device, 62% were deemed unsuitable for antico-
WATCHMAN device was FDA approved for patients with non- agulation and only 27% were treated with anticoagulation as
valvular AF in whom long-term anticoagulation is indicated, part of a post-implant antithrombotic therapy course. The rest
but who have an appropriate rationale to seek a nonpharma- of the patients were receiving either dual (59%) or single (7%)
cologic alternative to anticoagulation (Holmes 2014; Reddy antiplatelet therapy, or no antithrombotic therapy (6%). At the
2014). The post-implant antithrombotic regimen within these 1-year follow-up, the rate of ischemic stroke was 1.1%, showing
two trials consisted of warfarin plus aspirin (81–325 mg) for success even in patients with contraindications to anticoagu-
45 days, followed by clopidogrel plus aspirin for 6 months, fol- lation (Boersma 2017).
lowed by aspirin alone indefinitely. With this protocol, patients Although the U.S. guidelines do not comment on a recom-
being considered for WATCHMAN placement must be able to mended antithrombotic strategy, the ESC guidelines offer a
tolerate warfarin for at least 6 weeks after device implantation table outlining suggested antithrombotic strategies for the
and must not have an LAA clot at the time of device placement. WATCHMAN and Amulet occlusion devices (Table 11).
Most WATCHMAN implant centers in the United States follow
an antithrombotic protocol similar to the PROTECT AF and Surgical LAA Occlusion or Exclusion
PREVAIL trials. However, in patients with absolute contraindi- Surgical ligation or amputation is another, though more
cations to anticoagulation, LAA closure with the WATCHMAN invasive means to exclude the LAA and conceivably reduce
device has been evaluated. In the ASAP study, a post-procedure stroke risk. In most instances, surgical exclusion of the LAA
regimen of clopidogrel plus aspirin for 6 months (or for 1–3 is performed in patients already undergoing cardiac sur-
months in patients at very high risk of bleeding) followed by gery and is not typically a stand-alone procedure. Surgical

Table 11. Antithrombotic Therapy After LAA Occlusiona

Device
(patient type) Aspirin Oral Anticoagulant Clopidogrel Comments

WATCHMAN (low 75–325 mg/day Initiate warfarin after Initiate 75 mg/ Some centers do not
bleeding risk) indefinitely procedure (INR day when OAC withhold OAC at time
2–3) until 45 days discontinued, continue of procedure (no data
or continue until until 6 mo after the to support/deny this
adequate LAA sealingb procedure approach)
is confirmed by TEE.
DOAC is a possible
alternative

WATCHMAN (high 75–325 mg/day None 75 mg/day for 1–6 Clopidogrel often
bleeding risk) indefinitely mo while ensuring administered for
adequate LAA sealingb shorter time in very
high-risk situations

ACP/Amulet 75–325 mg/day None 75 mg/day for 1–6 Clopidogrel may


indefinitely mo while ensuring replace long-term
adequate LAA sealingb aspirin if better
tolerated

a
Note: Load aspirin or clopidogrel before procedure, if untreated. Heparin with activated clotting time > 250 s before or immediately
after transseptal puncture for all patients, followed by LMWH when warfarin is needed.
b
Less than 5-mm leak.
ACP = Amplatzer Cardiac Plug; DOAC = direct oral anticoagulant; LAA = left atrial appendage; OAC = oral anticoagulant;
TEE = transesophageal echocardiography.

PSAP 2022 Book 1 • Cardiology 140 Management of Atrial Fibrillation


exclusion of the LAA is performed in several ways. Epicardial of successful closure and the unknown impact of LAA occlu-
surgical techniques to exclude the LAA include simple suture sion on further thromboembolic events seem to underscore
ligation, oversewing the base without excision, excising the the need for continued anticoagulation after the procedure.
LAA and oversewing the base, and surgical stapling and Of note, in the LAAOS III trial, most patients continued OAC
excision. Endocardial surgical techniques include inversion after the procedure, with a total of 75.3% (LAA occlusion
of the appendage and amputation and then oversewing group) and 78.2% (no-occlusion group) of patients remain-
the base from the endocardial aspect (January 2014). The ing on OAC at the 3-year visit (Whitlock 2021). At a minimum,
FDA-approved AtriClip LAA Exclusion System is also an echocardiography should be performed after the operation to
option for mechanical surgical closure. confirm LAA closure before decisions are made regarding the
need for long-term anticoagulation.
Indications
Current guidelines state that surgical occlusion of the LAA can
CONTEMPORARY STRATEGIES
be considered in patients with AF undergoing cardiac surgery
FOR RHYTHM CONTROL
as a component of an overall heart team approach to man-
aging AF (IIB, B-NR) (January 2019). However, the guidelines Both catheter and surgical ablation procedures offer an alter-
suggest no specific means of surgical LAA closure, partly native to AAD therapy in the maintenance of sinus rhythm.
because of the tremendous diversity among surgical tech- The role of catheter and surgical ablation technologies has
niques, cardiac and atrial anatomies, and concomitant thera- rapidly been evolving, expanding the scope of these proce-
pies. As a result, studies of surgical LAA removal have long dures across a wide variety of patients with AF.
yielded inconsistent results and highly variable rates of suc-
cessful LAA occlusion and stroke reduction (January 2014). Catheter Ablation
Despite this, surgical LAA exclusion is still commonly per- Atrial fibrillation catheter ablation is performed by an elec-
formed as part of cardiac surgery in patients with AF. How- trophysiologist using either cryoballoon or irrigated radiof-
ever, the LAAOS III trial published in 2021 offers additional requency techniques. During AF ablation, a catheter is
evidence in support of surgical LAA occlusion during cardiac inserted percutaneously into the LA through transseptal
surgery. In this multicenter trial, 4770 patients with AF and a puncture. Energy in the form of either heat (radiofrequency)
CHA 2DS2-VASc score of at least 2 who were undergoing car- or extreme cold (cryoballoon) is delivered to the surround-
diac surgery for another indication were randomized to receive ing endocardial tissue to create linear lesions around the
either LAA occlusion or no occlusion. Patients were followed antrum of the PVs; these lesions form scar tissue that can-
for a mean of 3.8 years. Trial results showed significantly lower not conduct or propagate electrical impulses. In many
rates of stroke or systemic embolism in the LAA occlusion cases, electrical triggers for AF are located near and within
group (114 patients [4.8%]) than in the no-occlusion group (168 the PVs. As such, the goal of the ablation procedure is to
patients [7.0%]) (HR 0.67; 95% CI, 0.53–0.85; p=0.001). Further- achieve complete electrical isolation of the PVs from the
more, the incidence of perioperative bleeding, HF, or death LA, with subsequent maintenance of sinus rhythm. More
did not differ between trial groups. In the LAAOS III trial, LAA advanced ablation techniques have also been developed
occlusion was primarily performed by amputation and closure that target additional or alternative sites (non-pulmonary
(“cut and sew”) in over 50% of patients (Whitlock 2021). foci); however, these are typically reserved for ablation in
patients with persistent or longstanding persistent AF, and
Complications the benefit of targeting these extrapulmonary sites is still
Procedure-related complications of surgical LAA exclusion undetermined.
are uncommon. One recent large meta-analysis noted no sig-
nificant differences in postoperative complications or the Indications
need for reoperation for bleeding between patients receiving In general, AF catheter ablation is recommended as a
LAA exclusion and those not receiving LAA exclusion during second-line therapy after failure or intolerance of class I or
any cardiac surgery (Atti 2018). A bigger limitation to surgical class III AADs. Table 12 summarizes these recommendations,
LAA exclusion is that closure may be incomplete, with echo- which are based on the results of several randomized con-
cardiographic follow-up suggesting incomplete occlusion in trolled trials showing the superiority of AF catheter ablation
more than 50% of patients (Kanderian 2008). Incomplete sur- over AADs regarding freedom from recurrent arrhythmia or
gical LAA occlusion has strongly been associated with the improvement in symptoms, exercise capacity, and quality of
occurrence of thromboembolic events (January 2014). life after medication failure (Hindricks 2021).
Several factors play into whether to pursue AF catheter
Anticoagulation Management ablation, including type of AF (paroxysmal vs. persistent vs.
It is unknown whether anticoagulation should be continued longstanding persistent), degree of symptoms, presence of
after surgical exclusion of the LAA. The highly variable rates structural heart disease, risk of complications, and patient

PSAP 2022 Book 1 • Cardiology 141 Management of Atrial Fibrillation


Table 12. Summary of Recommendations for AF Catheter Ablation or Surgical Ablation to Maintain Sinus Rhythm

COR LOE Recommendation

Catheter AF Ablation

I A AF catheter ablation is useful in patients with symptomatic paroxysmal AF refractory to or intolerant of at


least one class I or III AAD when a rhythm-control strategy is desired

I C Before consideration of AF catheter ablation, assessment of the procedural risks and outcomes relevant to
the individual patient is recommended

IIa A AF catheter ablation is reasonable for some patients with symptomatic persistent AF refractory to or
intolerant of at least one class I or III AAD

IIa B In patients with recurrent symptomatic paroxysmal AF, catheter ablation is a reasonable initial rhythm-
control strategy before therapeutic trials of AAD therapy after weighing the risks and outcomes of drug and
ablation therapy

IIb B AF catheter ablation can be considered in patients with symptomatic longstanding (> 12 mo) persistent AF
refractory to or intolerant of at least one class I or III AAD when a rhythm-control strategy is desired

IIb B-R AF catheter ablation may be reasonable in selected patients with symptomatic AF and HFrEF to potentially
lower mortality rate and reduce hospitalization for HF

IIb C AF catheter ablation can be considered before initiation of a class I or III AAD in patients with symptomatic
persistent AF when a rhythm-control strategy is desired

III: Harm C AF catheter ablation should not be performed in patients who cannot be treated with anticoagulant therapy
during and after the procedure

III: Harm C AF catheter ablation to restore sinus rhythm should not be performed solely to obviate the need for
anticoagulation

Surgical AF Ablation

IIa C An AF surgical ablation procedure is reasonable for selected patients with AF undergoing cardiac surgery for
other indications

IIb B A stand-alone AF surgical ablation procedure may be reasonable for selected patients with highly
symptomatic AF not well managed with other approaches

AAD = antiarrhythmic drug; AF = atrial fibrillation; HFrEF = heart failure with reduced ejection fraction.
Information from: January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/ACC/HRS guideline
for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association
Task Force on Clinical Practice Guidelines and the Heart Rhythm Society in Collaboration with the Society of Thoracic Surgeons
[published correction appears in Circulation 2019;140:e285]. Circulation 2019;140:e125-e151; January CT, Wann LS, Alpert JS, et al.
2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of
Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society [published correction
appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

preference. Overall, the main clinical benefit of AF catheter In contrast, in patients with HFrEF and AF, randomized
ablation is reduction in arrhythmia-related symptoms. In the controlled trials have shown significant reductions in all-
recent CABANA trial, symptom improvement was confirmed cause mortality and hospitalization with AF catheter abla-
with catheter ablation compared with AAD therapy; however, tion. In the CASTLE-AF trial, 363 patients with HFrEF (EF less
an ablation strategy did not significantly reduce the primary than 35%) and symptomatic AF were randomized to either
composite outcome of death, disabling stroke, serious bleed- catheter ablation or medical therapy (rate or rhythm control).
ing, or cardiac arrest compared with AAD therapy (Packer After a median follow-up of 37.8 months, the primary compos-
2019). As such, until a randomized controlled trial shows an ite end point of death from any cause or hospitalization for
improvement in clinical end points outside symptoms, AF worsening HF was significantly lower in the catheter ablation
catheter ablation in the general AF population is typically group (28.5%) than in the medical therapy group (44.6%) (Mar-
reserved for symptom relief and is not indicated in asymp- rouche 2018). Given these findings, new recommendations
tomatic patients. were added to the 2019 AHA/ACC/HRS guidelines supporting

PSAP 2022 Book 1 • Cardiology 142 Management of Atrial Fibrillation


Table 13. Procedure-Related Complications in Catheter Ablation of AF

Complication Complication
Severity Complication Type Rate (%) Treatment

Life threatening Periprocedural death < 0.1 N/A

Esophageal perforation/fistula < 0.5 CT or MRI of esophagus, avoiding endoscopy,


immediate surgical correction

Periprocedural thromboembolic event < 1.0 Consider lysis therapy

Cardiac tamponade ~1 Pericardiocentesis, emergency surgical drainage

Severe PV stenosis < 1.0 PV dilation/stent or no therapy

Persistent phrenic nerve palsy < 1.0 None, usually resolves spontaneously

Vascular access complications 2–4 Observation, compression, possible surgical


intervention

Moderate or minor Various: 1–2 MI: Standard therapy


• MI Pericarditis: NSAIDs, colchicine, steroids
• Pericarditis Radiation injury: Treat as burn injury
• Radiation injury

Complications Asymptomatic cerebral embolism 5–15 N/A


of unknown
significance

PV = pulmonary vein.
Information from: Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of atrial fibrillation
developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J 2021;42:373-498;
January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report
of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm
Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76.

AF ablation in patients with HFrEF and symptomatic AF to rather than using early repeat ablation, it is usually reason-
potentially reduce mortality and HF hospitalizations (January able to try an AAD for additional rhythm control or cardiover-
2019). sion for persistent AF. Theoretically, aggressive treatment
of early AF recurrences helps prevent both structural and
Complications and Recurrence electrical remodeling and improves long-term outcomes,
Although AF catheter ablation is relatively safe when per- but studies are needed to clarify the optimal management
formed in experienced centers, several important risks and strategy. In later AF recurrences (i.e., after 3 months), recur-
complications are associated with it, as summarized in rence is usually an indication of PV conduction recovery,
Table  13. Catheter ablation may be complicated by peripro- and repeat ablation or AAD initiation may be helpful (Janu-
cedural stroke, resulting from catheter manipulation and the ary 2014).
creation of lesions in the endocardial tissue (Basu-Ray 2021).
Around 1% of patients who receive AF ablation are at risk of Anticoagulation Management
stroke, and post-ablation brain MRI reveals silent stroke in Anticoagulation plays an integral role in both the peri- and
14%–40% of patients (Mao 2020). postprocedural management of AF catheter ablation, so
Alternatively, as a result of background anticoagulation, much so that patients who cannot be treated with anticoag-
use of large sheaths, and femoral access, bleeding compli- ulation during and after the procedure should not undergo
cations can occur and be severe. Most complications occur AF catheter ablation. Large areas of LA endothelium are left
within the first 24 hours after the procedure; however, some damaged from the procedure and can incite thrombus forma-
may appear 1–2 months later (Arbelo 2017, 2014, 2012). tion, as can manipulation of vascular sheaths and catheters.
Recurrences of AF after a catheter ablation are common After the operation, stunning of atrial tissue may occur, last-
during the first 3 months and do not rule out long-term pro- ing for weeks or even months and contributing to blood stasis
cedural success, though they can increase the risk of rehos- and thrombus formation.
pitalization. In early AF recurrences (i.e., within 3 months),

PSAP 2022 Book 1 • Cardiology 143 Management of Atrial Fibrillation


Table 14. Periprocedural Anticoagulation Strategies for AF Catheter Ablation

COR LOE Recommendation

Before Ablation

I A For patients undergoing AF catheter ablation who have been therapeutically anticoagulated with warfarin or
dabigatran, performance of the procedure without interruption of anticoagulation is recommended

I B-R For patients undergoing AF catheter ablation who have been therapeutically anticoagulated with rivaroxaban,
performance of the procedure without interruption of rivaroxaban is recommended

IIa B-NR For patients undergoing AF catheter ablation who have been therapeutically anticoagulated with a DOAC
other than dabigatran or rivaroxaban, performance of the procedure without interruption of anticoagulation is
reasonable

IIa B-NR For patients anticoagulated with a DOAC before AF catheter ablation, it is reasonable to hold one or two doses
of the DOAC before AF ablation with reinitiation after ablation

During Ablation

I B-NR UFH should be administered before or immediately after transseptal puncture during AF catheter ablation
procedures and adjusted to achieve and maintain an ACT of at least 300 s

IIa B-NR Administration of protamine after AF catheter ablation to reverse UFH is reasonable

After Ablation

I C-EO In patients who do not undergo therapeutic anticoagulation before AF catheter ablation and in whom warfarin
will be used for anticoagulation after ablation, LMWH or IV UFH should be used as a bridge for initiation of
systemic anticoagulation with warfarin after AF ablation

I C-EO Systemic anticoagulation with warfarin or a DOAC is recommended for at least 2 mo after AF catheter ablation

I C-EO Adherence to AF anticoagulation guidelines is recommended for patients who have undergone an AF ablation
procedure, regardless of the apparent success or failure of the procedure

I C-EO Decisions regarding continuation of systemic anticoagulation > 2 mo after ablation should be based on the
patient’s stroke risk and not on the perceived success or failure of the ablation procedure

IIa C-EO In patients who have not been receiving anticoagulation before AF catheter ablation or in whom
anticoagulation with a DOAC or warfarin has been interrupted before ablation, administration of a DOAC 3–5
hr after achievement of hemostasis is reasonable after ablation

ACT = activated clotting time; AF = atrial fibrillation; DOAC = direct oral anticoagulant; HFrEF = heart failure with reduced ejection frac-
tion; LMWH = low-molecular-weight heparin; UFH = unfractionated heparin.
Information from: Calkins H, Hindricks G, Cappato R, et al. 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement
on catheter and surgical ablation of atrial fibrillation. Heart Rhythm 2017;14:e275-e444.

Because of the considerable risks of both bleeding in major or minor bleeding, pericardial tamponade, pericardial
and thromboembolic complications of the ablation proce- effusion, or puncture complications; however, uninterrupted
dure itself, there are challenges in determining the optimal anticoagulation resulted in significantly fewer silent strokes
periprocedural anticoagulation strategy for patients. The lat- than interrupted anticoagulation (Basu-Ray 2021; Mao 2020).
est international guidelines include detailed recommenda- Post-procedure anticoagulation should be continued for at
tions for anticoagulation surrounding the ablation procedure least 2 months after ablation in all patients. Afterward, the
(before, during, and after ablation), as summarized in Table 14 decision to continue OAC is largely based on stroke risk rather
(Calkins 2017). than rhythm status (Hindricks 2021).
Since publication of these guidelines, additional evidence
has been published in support of uninterrupted anticoagula- Surgical Ablation
tion with DOACs. Two large meta-analyses comparing patients Unlike catheter ablation procedures that create lesions on the
receiving uninterrupted anticoagulation with interrupted anti- endocardial surface of the LA, surgical ablation techniques
coagulation during AF catheter ablation noted no differences use an epicardial approach. In most cases, surgical ablations

PSAP 2022 Book 1 • Cardiology 144 Management of Atrial Fibrillation


Patient Care Scenario
H.J. is a 70-year-old woman who underwent AF catheter 410 milliseconds and an LVEF of > 55%. Which one of
ablation about 4 weeks ago. Her medical history includes the following interventions would be most reasonable
hypertension, hyperlipidemia, and osteoarthritis. Her for H.J.?
home drugs include rivaroxaban 20 mg daily, verapamil A. Initiate dofetilide 500 mcg orally twice daily.
extended-release 180 mg daily, amlodipine 10 mg B. Initiate flecainide 100 mg orally twice daily.
daily and atorvastatin 20 mg daily. An ECG and a TEE
C. Increase verapamil to 240 mg once daily.
performed during follow-up today in clinic reveal AF
D. Refer for repeat AF catheter ablation.
(ventricular rate 75 beats/minute) with a QTc interval of
ANSWER
Early recurrences (within 3 months) of AF after catheter for persistent AF (Answer C is incorrect). Both flecainide
ablation are common and are not usually indicative of pro- and dofetilide are AADs that may be considered for H.J;
cedural success (Answer D is incorrect); however, they can however, dofetilide requires hospitalization for initiation
increase risk of rehospitalization. In early recurrences, and is contraindicated with concomitant use of verapamil
guidelines recommend that is generally reasonable to (Answer A is incorrect, Answer B is correct).
trial an AAD for additional rhythm control or cardioversion

1. Hindricks G, Potpara T, Dagres N, et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collab-
oration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J. 2021;42(5):373-498.
2. January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management
of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical
Practice Guidelines and the Heart Rhythm Society in Collaboration With the Society of Thoracic Surgeons [published correction appears
in Circulation. 2019 Aug 6;140(6):e285]. Circulation. 2019;140(2):e125-e151.

are performed by a cardiothoracic surgeon in patients who Complications


are undergoing cardiac surgery for another reason (e.g., cor- Many complications are possible during surgical AF ablation
onary bypass grafting, valve repair or replacement). The prin- that largely depend on patient characteristics, procedure
cipal goal for surgical AF ablation is the same as for catheter approach (open sternotomy vs. minimally invasive thoracot-
AF ablation – to create tissue lesions that result in conduc- omy), procedure type (lesion patterns), and concomitant car-
tion block of micro- and macro-reentrant circuits and main- diac surgeries (valve replacement/repair or coronary bypass
tain normal sinus rhythm. This creates a “maze” of functional grafting). Although the risk of periprocedural stroke also
myocardium within the atrium that allows for appropriate exists with surgical AF ablation, risk is much lower than with
propagation of atrial depolarization. catheter ablation because of the lack of transseptal punc-
Surgical ablation techniques have evolved significantly over ture and endocardial catheter manipulation. The risk of PV
the past 30 years. Today, the most common surgical ablation stenosis is also much lower with a surgical approach. On the
procedure is known as the Cox maze IV and is considered the contrary, given the large extent of tissue damaged in the cre-
gold standard of surgical ablation techniques. The Cox maze ation of epicardial lesions, surgical ablation is associated
IV consists of a specific pattern of linear scars made on the with a higher incidence of atrial dysfunction and diminished
epicardial surface using incision, radiofrequency, or cryoab- atrial contractility compared with catheter ablation (Buber
lation. In many cases, the LAA is also surgically removed or 2011). In addition, the postoperative need for a permanent
ligated during these procedures (January 2014). pacemaker is higher with surgical ablation, especially with
bi-atrial lesion sets, though published results are highly vari-
Indications able and incidence ranges from 6.8% to 21.5% (Hindricks
Although AF surgical ablation has been shown to be effica- 2021).
cious in restoring and maintaining sinus rhythm, long-term
clinical outcomes such as quality of life, stroke, hospitaliza- Anticoagulation Management
tion, and mortality have not been well established. The ACC/ Few to no data are available regarding the optimal approach
AHA 2014 guidelines give a weak recommendation for consid- for anticoagulation after AF surgical ablation. Despite the
eration of AF surgical ablation in patients with AF undergoing absence of evidence, the 2017 international expert consen-
cardiac surgery for other indications, or as a stand-alone pro- sus guidelines recommend that the decisions to provide
cedure for selected highly symptomatic patients with AF for anticoagulation and the therapy duration be made on an
whom other approaches have failed (i.e., AADs, direct current individual basis, with consideration for the patient’s stroke
cardioversion [DCCV], catheter ablation) (see Table 12) (Janu- risk and bleeding risk and whether the LAA was ligated.
ary 2014). Authors state that anticoagulation may be reasonable in

PSAP 2022 Book 1 • Cardiology 145 Management of Atrial Fibrillation


patients for several months after surgical ablation with low Indications and Complications
bleeding risk. For patients in whom LAA closure or ligation Hybrid AF procedures have been developed and refined over
was performed at the time of surgical ablation, TEE-based the past several years but are not yet formally indicated by
assessment of LAA closure should be performed when the current AHA/ACC guidelines. The 2020 ESC AF guide-
anticoagulation discontinuation is being considered (Calk- lines and the 2017 international consensus guidelines offer
ins 2017). a weak recommendation in support of a hybrid AF ablation
in patients with symptomatic paroxysmal or persistent AF
Hybrid Surgical/Catheter Ablation refractory to AAD therapy for whom at least one attempt at
Over the past decade, a hybrid approach to surgical and cath- catheter AF ablation has failed or for those who have signif-
eter AF ablation has been developed whereby a minimally icant risk factors for AF recurrence (Hindricks 2021; Calkins
invasive epicardial surgical ablation performed by a cardio- 2017). Hybrid AF ablation procedures achieve significantly
thoracic surgeon is combined with a percutaneous cathe- higher rates of long-term freedom from AF than AF cathe-
ter ablation performed by an electrophysiologist. This hybrid ter ablation, though they are associated with more compli-
approach can be performed as a single intervention, where cations (13.8% vs. 5.9%) (van der Heijden 2019). Although
both procedures are performed during the same hospitaliza- complications are decreasing with procedure refinement and
tion, or sequentially, where surgical and catheter ablation pro- increased center experiences, they may involve phrenic nerve
cedures are separated by no more than 6 months. Although palsy, pericardial effusion or tamponade, esophageal injury
evidence is lacking to support this practice, an LAA clip (Atri- or perforation, bleeding, or stroke/TIA.
Clip) may be included in a hybrid AF ablation, and an AAD can
Anticoagulation Management
also be considered after ablation to increase potential treat-
Much like surgical AF ablation, there are few to no data to
ment success.
recommend or refute anticoagulation after a hybrid AF

Table 15. Summary of Recommendations for Specific Patient Groups and AF

COR LOE Recommendation

Hypertrophic Cardiomyopathy

I B Anticoagulation is indicated in HCM with AF independent of the CHA 2DS2-VASc score

IIa C AADs can help prevent recurrent AF in HCM. Amiodarone or disopyramide combined with a β-blocker
or non-DHP CCB is reasonable

IIa B AF catheter ablation can be beneficial for HCM to facilitate a rhythm-control strategy when AADs fail
or are not tolerated

IIb C Sotalol, dofetilide, and dronedarone can be considered for a rhythm-control strategy in HCM

AF Complicating ACS

I B-R With ACS and AF with a CHA 2DS2-VASc score ≥ 2, anticoagulation is recommended unless CI

I C Urgent DCCV of new-onset AF in the setting of ACS is recommended for patients with hemodynamic
compromise, ongoing ischemia, or inadequate rate control

I C IV β-blockers are recommended to slow RVR with ACS and no HF, hemodynamic instability,
bronchospasm

IIa B-NR If triple therapy (anticoagulant, ASA, and P2Y12 inhibitor) is prescribed for patients with AF at
increased risk of stroke (CHA 2DS2-VASc score ≥ 2) who have undergone PCI with stenting for ACS, it
is reasonable to choose clopidogrel in preference to prasugrel

IIa B-R In patients with AF and CHA 2DS2-VASc score ≥ 2 who have undergone PCI with stenting for ACS,
double therapy with a P2Y12 inhibitor (clopidogrel or ticagrelor) and dose-adjusted warfarin is more
reasonable to reduce risk of bleeding than triple therapy

IIa B-R In patients with AF and CHA 2DS2-VASc score ≥ 2 who have undergone PCI with stenting for ACS,
double therapy with P2Y12 inhibitors (clopidogrel) and low-dose rivaroxaban 15 mg daily is more
reasonable to reduce risk of bleeding than triple therapya

(continued)

PSAP 2022 Book 1 • Cardiology 146 Management of Atrial Fibrillation


Table 15. Summary of Recommendations for Specific Patient Groups and AF  (continued)

COR LOE Recommendation

IIa B-R In patients with AF and CHA 2DS2-VASc score ≥ 2 who have undergone PCI with stenting for ACS,
double therapy with P2Y12 inhibitors (clopidogrel) and dabigatran 150 mg BID is more reasonable to
reduce risk of bleeding than triple therapy

IIb B-R If triple therapy (anticoagulant, ASA, and P2Y12 inhibitor) is prescribed for patients with AF who are
at increased risk of stroke (CHA 2DS2-VASc score ≥ 2) and who have undergone PCI with stenting for
ACS, changing to double therapy (anticoagulant and P2Y12 inhibitor) at 4–6 wk can be considered

IIb C Amiodarone or digoxin can be considered to slow RVR with ACS and AF and severe LV dysfunction
and HF or hemodynamic instability

IIb C Non-DHP CCBs can be considered to slow RVR with ACS and AF only in the absence of significant HF
or hemodynamic instability

Hyperthyroidism

I C β-Blockers are recommended to control ventricular rate with AF complicating thyrotoxicosis unless CI

I C When β-blockers cannot be used, a non-DHP CCB is recommended to control ventricular rate

Pulmonary Diseases

I C A non-DHP CCB is recommended to control ventricular rate with AF and COPD

I C Cardioversion should be tried for patients with pulmonary disease who become hemodynamically
unstable with new-onset AF

WPW and Preexcitation Syndromes

I C Cardioversion is recommended for patients with AF, WPW syndrome, and RVR who are
hemodynamically compromised

I C IV procainamide or ibutilide to restore sinus rhythm or slow ventricular rate is recommended for
patients with preexcited AF and RVR who are not hemodynamically compromised

I C Catheter ablation of the accessory pathway is recommended in symptomatic patients with preexcited
AF, especially if the accessory pathway has a short refractory period

III: Harm B Use of IV amiodarone, adenosine, digoxin, or non-DHP CCBs in patients with WPW syndrome who
have preexcited AF is potentially harmful

Heart Failure

I B A β-blocker or non-DHP CCB is recommended for persistent or permanent AF in patients with HFpEF

I B In the absence of preexcitation, an IV β-blocker (or a non-DHP CCB with HFpEF) is recommended to
slow ventricular response to AF in the acute setting, with caution in patients with overt congestion,
hypotension, or HFrEF

I B In the absence of preexcitation, IV digoxin or amiodarone is recommended to control HR acutely

I C Assess HR during exercise and adjust pharmacologic treatment in symptomatic patients during
activity

I C Digoxin is effective to control resting HR with HFrEF

IIa B A combination of digoxin and a β-blocker (or a non-DHP CCB with HFpEF) is reasonable to control
resting and exercise HR with AF

IIa B It is reasonable to perform AV node ablation with ventricular pacing to control HR when
pharmacologic therapy is insufficient or not tolerated

IIa C IV amiodarone can be useful to control HR with AF when other measures are unsuccessful or CI

(continued)

PSAP 2022 Book 1 • Cardiology 147 Management of Atrial Fibrillation


Table 15. Summary of Recommendations for Specific Patient Groups and AF  (continued)

COR LOE Recommendation

IIa B With AF and RVR causing or suspected of causing tachycardia-induced cardiomyopathy, it is


reasonable to use a rhythm-control strategy

IIa C In patients with chronic HF who remain symptomatic from AF despite a rate-control strategy, it is
reasonable to use a rhythm-control strategy

IIb C Amiodarone can be considered when resting and exercise HR cannot be controlled with a β-blocker
(or a non-DHP CCB with HFpEF) or digoxin, alone or in combination

IIb C AV node ablation can be considered when rate cannot be controlled and tachycardia-mediated
cardiomyopathy is suspected

III: Harm C AV node ablation should not be performed without a pharmacologic trial to control ventricular rate

III: Harm C For rate control, IV non-DHP CCBs, IV β-blockers, and dronedarone should not be administered with
decompensated HF

Postoperative Cardiac and Thoracic Surgery

I A A β-blocker is recommended to treat postoperative AF unless CI

I B A non-DHP CCB is recommended when a β-blocker is inadequate to achieve rate control with
postoperative AF

IIa A Preoperative amiodarone reduced AF with cardiac surgery and is reasonable as prophylactic therapy
for patients at high risk of postoperative AF

IIa B It is reasonable to restore sinus rhythm pharmacologically with ibutilide or DCCV with postoperative
AF

IIa B It is reasonable to administer an AAD to maintain sinus rhythm with recurrent or refractory
postoperative AF

IIa B It is reasonable to administer antithrombotic medications for postoperative AF

IIa C It is reasonable to manage new-onset postoperative AF with rate control and anticoagulation with
cardioversion if AF does not revert spontaneously to sinus rhythm during follow-up

IIb B Prophylactic sotalol can be considered for patients at risk of AF after cardiac surgery

IIb B Colchicine can be considered after the operation to reduce AF after cardiac surgery

a
Apixaban 5 mg BID can also be considered, given the results of AUGUSTUS trial (Lopes RD, Heizer G, Aronson R, et al. Antithrombotic
therapy after acute coronary syndrome or PCI in atrial fibrillation. N Engl J Med 2019;380:1509-24) (published after 2019 AHA/ACC/
HRS guideline update).
ACS = acute coronary syndrome; ASA = aspirin; CI = contraindicated; DCCV = direct current cardioversion; HCM = hypertrophic
cardiomyopathy; HR = heart rate; PCI = percutaneous coronary intervention; WPW = Wolff-Parkinson-White.
Information from: January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial
fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the
Heart Rhythm Society [published correction appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll Cardiol 2014;64:e1-e76;
January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/ACC/HRS guideline for the
management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task
Force on Clinical Practice Guidelines and the Heart Rhythm Society in Collaboration with the Society of Thoracic Surgeons
[published correction appears in Circulation 2019;140:e285]. Circulation 2019;140:e125-e151.

ablation, and decisions surrounding anticoagulation initia- AF MANAGEMENT IN SPECIAL


tion or withdrawal should be made on the basis of patient- POPULATIONS
and procedure-specific factors. Although great strides have been made in managing AF as
a whole, many specific patient subgroups warrant additional

PSAP 2022 Book 1 • Cardiology 148 Management of Atrial Fibrillation


Practice Points
• AF is a CV pandemic and contributes to signifi- • Pharmacists should recognize that both stroke
cant patient morbidity and mortality. The patho- risk and a patient’s preferred AF treatment strat-
physiology of AF is complex, and both pharma- egy often change and may evolve over time. In
cologic and nonpharmacologic strategies for AF addition, management of concomitant disease
treatment have evolved tremendously over the states and stroke risk factors is an important part
past several years. Pharmacists should have a of an AF treatment strategy.
sound understanding of AF as a disease state, • Contemporary nonpharmacologic strategies for
together with an understanding of the currently stroke prevention in AF include percutaneous
available treatment modalities, in order to make LAA occlusion or ligation, or surgical LAA exclu-
appropriate recommendations surrounding sion or ligation. Pharmacists should be aware of
pharmacotherapy that maximizes benefit while these procedures, potential complications, and
minimizing harm. how anticoagulant therapy may be used in these
• Stroke prevention is a cornerstone of therapy in patients.
AF management, regardless of treatment strat- • Contemporary nonpharmacologic strategies for
egy, and is quantified by the CHA2DS2-VASc risk rhythm control in AF include catheter ablation,
assessment tool. In determining appropriate an- surgical ablation, or a hybrid surgical/catheter
ticoagulant therapy, pharmacists should assess ablation. Pharmacists should be aware of these
stroke and bleeding risk, patient comorbidities, procedures, potential complications, and how
indications/contraindications, renal/hepatic func- AAD and anticoagulant therapy can be used in
tion, potential for interacting medications, adher- these patients.
ence, and affordability at a minimum. • Certain populations of patients with AF warrant
• Symptom management of AF may involve either special consideration because concomitant
a rate- or rhythm-control strategy, depending on disease states (e.g., ACS, hyperthyroidism,
patient-specific factors and shared decision-mak- congestive heart failure) may dictate alternative
ing. Pharmacists should be knowledgeable about treatment strategies that differ from general
which medications are appropriate for each AF guideline recommendations. Pharmacists
strategy as well as consider comorbidities, indi- should be familiar with these subgroups and
cations/contraindications, renal/hepatic function, how AF pharmacotherapy decisions may be
potential for interacting medications, adherence, affected as a result.
and affordability.

consideration when treating concomitant AF. Pharmacists from the ESC-EHRA atrial fibrillation ablation long-term
should be cognizant of these patient AF subgroups and aware registry. Eur Heart J 2017;38:1303-16.
of potential caveats to consider regarding appropriate phar- Arbelo E, Brugada J, Hindricks G, et al. ESC-EURObserva-
macotherapy and disease state management. These condi- tional Research Programme: the Atrial Fibrillation Abla-
tions are summarized in Table 15 and discussed elsewhere in tion Pilot Study, conducted by the European Heart Rhythm
this module in greater detail. Association. Europace 2012;14:1094-103.

Arbelo E, Brugada J, Hindricks G, et al. The atrial fibrillation


CONCLUSION ablation pilot study: a European survey on methodology
Much progress has been made in preventing, detecting, and results of catheter ablation for atrial fibrillation con-
ducted by the European Heart Rhythm Association. Eur
and treating AF, with much more to come. In the aging pop-
Heart J 2014;35:1466-78.
ulation, the global burden of AF is increasing, and it has
become one of the most common disease states world- Atti V, Anantha-Narayanan M, Turagam MK, et al. Surgical
wide. Pharmacists can play an integral role in patient edu- left atrial appendage occlusion during cardiac surgery:
cation and treatment surrounding AF, and an understanding a systematic review and meta-analysis. World J Cardiol
2018;10:242-9.
of contemporary nonpharmacologic AF management strat-
egies is essential for making appropriate pharmacotherapy Basu-Ray I, Khanra D, Kupó P, et al. Outcomes of uninter-
recommendations. rupted vs interrupted periprocedural direct oral antico-
agulants in atrial fibrillation ablation: a meta-analysis.
J Arrhythm 2021;37:384-93.
REFERENCES
Arbelo E, Brugada J, Blomstrom-Lundqvist C, et al. Contem- Boersma LV, Ince H, Kische S, et al. Efficacy and safety of
porary management of patients undergoing atrial fibril- left atrial appendage closure with WATCHMAN in patients
lation ablation: in-hospital and 1-year follow-up findings with or without contraindication to oral anticoagulation:

PSAP 2022 Book 1 • Cardiology 149 Management of Atrial Fibrillation


1-year follow-up outcome data of the EWOLUTION trial. Kanderian AS, Gillinov AM, Pettersson GB, et al. Success
Heart Rhythm 2017;14:1302-8. of surgical left atrial appendage closure: assessment by
transesophageal echocardiography. J Am Coll Cardiol
Boersma LV, Schmidt B, Betts TR, et al. Implant success and 2008;52:924-9.
safety of left atrial appendage closure with the WATCH-
MAN device: peri-procedural outcomes from the EWOLU- Kirchhof P, Camm AJ, Goette A, et al. Early rhythm-control
TION registry. Eur Heart J 2016;37:2465. therapy in patients with atrial fibrillation. N Engl J Med
2020;383:1305.
Buber J, Luria D, Sternik L, et al. Left atrial contractile func-
tion following a successful modified maze procedure Kotecha D, Bunting KV, Gill SK, et al. Effect of digoxin vs
at surgery and the risk for subsequent thromboembolic bisoprolol for heart rate control in atrial fibrillation on
stroke. J Am Coll Cardiol 2011;58:1614-21. patient-reported quality of life: the RATE-AF randomized
clinical trial. JAMA 2020;324:2497-508.
Calkins H, Hindricks G, Cappato R, et al. 2017 HRS/EHRA/
ECAS/APHRS/SOLAECE expert consensus statement on Lin HJ, Wolf PA, Kelly-Hayes M, et al. Stroke severity in atrial
catheter and surgical ablation of atrial fibrillation. Heart fibrillation. The Framingham Study. Stroke 1996;27:1760-4.
Rhythm 2017;14:e275-e444.
Lip GYH. The ABC pathway: an integrated approach to
Cappato R, Ezekowitz MD, Klein AL, et al. Rivaroxaban vs. improve AF management. Nat Rev Cardiol 2017;14:627-8.
vitamin K antagonists for cardioversion in atrial fibrilla-
tion. Eur Heart J 2014;35:3346-55. Løfgren B, Pareek M, Larsen JM. Uninterrupted dabigatran
versus warfarin for ablation in atrial fibrillation. N Engl J
Chung MK, Refaat M, Shen WK, et al. Atrial fibrillation: Med 2017;377:494-5.
JACC Council perspectives. J Am Coll Cardiol
2020;75:1689-713. Mao YJ, Wang H, Huang PF. Peri-procedural novel oral
anticoagulants dosing strategy during atrial fibrillation
Ezekowitz MD, Pollack CV Jr, Halperin JL, et al. Apixaban ablation: a meta-analysis. Pacing Clin Electrophysiol
compared to heparin/vitamin K antagonist in patients 2020;43:1104-14.
with atrial fibrillation scheduled for cardioversion: the
EMANATE trial. Eur Heart J 2018;39:2959-71. Marrouche NF, Brachmann J, Andresen D, et al. Catheter
ablation for atrial fibrillation with heart failure. N Engl J
Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines Med 2018;378:417-27.
for the diagnosis and management of atrial fibrillation
developed in collaboration with the European Associa- Marzona I, Proietti M, Farcomeni A, et al. Sex differences in
tion for Cardio-Thoracic Surgery (EACTS). Eur Heart J stroke and major adverse clinical events in patients with
2021;42:373-498. atrial fibrillation: a systematic review and meta-analysis of
993,600 patients. Int J Cardiol 2018;269:182-91.
Holmes DR Jr, Doshi SK, Kar S, et al. Left atrial appendage
closure as an alternative to warfarin for stroke prevention Nielsen PB, Overvad TF. Female sex as a risk modifier for
in atrial fibrillation: a patient-level meta-analysis. J Am stroke risk in atrial fibrillation: using CHA2DS2-VASc
Coll Cardiol 2015;65:2614-23. versus CHA2DS2-VA for stroke risk stratification in
atrial fibrillation: a note of caution. Thromb Haemost
Holmes DR Jr, Kar S, Price MJ, et al. Prospective randomized 2020;120:894-8.
evaluation of the WATCHMAN Left Atrial Appendage Clo-
sure Device in patients with atrial fibrillation versus long- Packer DL, Mark DB, Robb RA, et al. Effect of catheter abla-
term warfarin therapy: the PREVAIL trial. J Am Coll Cardiol tion vs antiarrhythmic drug therapy on mortality, stroke,
2014;64:1-12. bleeding, and cardiac arrest among patients with atrial
fibrillation: the CABANA randomized clinical trial. JAMA
January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS 2019;321:1261-74.
guideline for the management of patients with atrial fibril-
lation: a report of the American College of Cardiology/ Pastori D, Farcomeni A, Pignatelli P, et al. ABC (Atrial
American Heart Association Task Force on Practice Guide- fibrillation Better Care) pathway and healthcare costs
lines and the Heart Rhythm Society [published correction in atrial fibrillation: the ATHERO-AF study. Am J Med
appears in J Am Coll Cardiol 2014;64:2305-7]. J Am Coll 2019a;132:856-61.
Cardiol 2014;64:e1-e76. Pastori D, Pignatelli P, Menichelli D, et al. Integrated care
January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS management of patients with atrial fibrillation and risk of
focused update of the 2014 AHA/ACC/HRS guideline for cardiovascular events: the ABC (Atrial fibrillation Better
the management of patients with atrial fibrillation: a report Care) pathway in the ATHERO-AF study cohort. Mayo Clin
of the American College of Cardiology/American Heart Proc 2019b;94:1261-7.
Association Task Force on Clinical Practice Guidelines and Ponamgi SP, Siontis KC, Rushlow DR, et al. Screening and
the Heart Rhythm Society in Collaboration with the Soci- management of atrial fibrillation in primary care. BMJ
ety of Thoracic Surgeons [published correction appears in 2021;373:n379.
Circulation 2019;140:e285]. Circulation 2019;140:e125-e151.
Proietti M, Romiti GF, Olshansky B, et al. Improved outcomes
by integrated care of anticoagulated patients with atrial

PSAP 2022 Book 1 • Cardiology 150 Management of Atrial Fibrillation


fibrillation using the simple ABC (Atrial fibrillation Better van der Heijden CAJ, Vroomen M, Luermans JG, et al. Hybrid
Care) pathway. Am J Med 2018;131:1359-1366.e6. versus catheter ablation in patients with persistent and
longstanding persistent atrial fibrillation: a system-
Reddy VY, Möbius-Winkler S, Miller MA, et al. Left atrial atic review and meta-analysis. Eur J Cardiothorac Surg
appendage closure with the WATCHMAN device in 2019;56:433-43.
patients with a contraindication for oral anticoagulation:
the ASAP study (ASA Plavix Feasibility Study with Watch- Whitlock RP, Belley-Cote EP, Paparella D, et al. Left atrial
man Left Atrial Appendage Closure Technology). J Am Coll appendage occlusion during cardiac surgery to prevent
Cardiol 2013;61:2551. stroke. N Engl J Med 2021;384:2081-91.

Reddy VY, Sievert H, Halperin J, et al. Percutaneous left Yoon M, Yang PS, Jang E, et al. Improved population-based
atrial appendage closure vs warfarin for atrial fibrillation: a clinical outcomes of patients with atrial fibrillation by
randomized clinical trial. JAMA 2014;312:1988-98. compliance with the Simple ABC (Atrial Fibrillation Better
Care) pathway for integrated care management: a nation-
Rienstra M, Hobbelt AH, Alings M, et al. Targeted therapy wide cohort study. Thromb Haemost 2019;119:1695-703.
of underlying conditions improves sinus rhythm mainte-
nance in patients with persistent atrial fibrillation: results
of the RACE 3 trial. Eur Heart J 2018;39:2987-96.

PSAP 2022 Book 1 • Cardiology 151 Management of Atrial Fibrillation


Self-Assessment Questions
Questions 1 and 2 pertain to the following case. 4. Which one of the following is best to recommend for pre-
R.C. is a 72-year-old woman (weight 75 kg) who presents to vention of thromboembolism for M.E.?
her cardiologist’s office with a 1-week history of fatigue, with
A. Apixaban 5 mg orally twice daily
periodic dizziness and palpitations. Her medical history is
B. Rivaroxaban 15 mg orally once daily with the
significant for hypertension, diabetes, and stage 3 chronic
evening meal
kidney disease (estimated CrCl 35 mL/minute/1.73 m2). R.C.
C. Anticoagulation not indicated
takes lisinopril 20 mg daily, furosemide 40 mg daily, insu-
D. Aspirin 81 mg orally once daily
lin glargine 15 units subcutaneously once daily, and insulin
aspart 5 units subcutaneously three times daily with meals.
Questions 5 and 6 pertain to the following case.
Her vital signs are stable, and a physical examination reveals
no signs of volume overload. An ECG reveals atrial fibrillation G.K. is a 82-year-old man (weight 58 kg) with a medical history
(AF) (no evidence of preexcitation). that includes mitral stenosis (moderate), hypertension, diabe-
tes, and coronary artery disease (CAD). He arrives at the ED
1. According to her CHA 2DS2-VASc score, which one of the
after 1 week of dizziness and palpitations. G.K. has had peri-
following best evaluates R.C.’s annual stroke risk?
odic episodes of “passing out” over the past 24 hours, accord-
A. 1.3% ing to his wife. In the ED, his blood pressure is 85/63 mm Hg
B. 2.2% and heart rate is 155 beats/minute. After his vital signs are
C. 3.2% obtained, G.K. begins to lose consciousness.
D. 4.0%
5. Which one of the following is best to recommend for
2. After a conversation between R.C. and her physician, a G.K.?
decision is made to pursue a rate-control strategy. Which
A. Administer amiodarone 150 mg intravenously
one of the following is best to recommend for R.C.?
administered over 10 minutes, followed by a
A. Diltiazem 180 mg extended release orally once daily continuous infusion at 1 mg/minute.
B. Amiodarone 400 mg orally twice daily for 2 weeks, B. Administer diltiazem 0.25 mg/kg intravenously
then 200 mg once daily administered over 2 minutes, followed by a
C. Digoxin 125 mcg orally daily continuous infusion at 5–15 mg/hour.
D. Flecainide 50 mg orally twice daily C. Administer therapeutic-dose low-molecular-weight
(LMWH) and perform an immediate TEE to assess
Questions 3 and 4 pertain to the following case. for LA thrombus.
M.E. is a 52-year-old woman (weight 68 kg) who presents to D. Provide immediate DCCV.
the ED this afternoon after new-onset palpitations and light- 6. Which one of the following is best to recommend to
headedness that began 3 hours ago after lunch with her reduce G.K.’s stroke risk?
neighbor. She says she has never had symptoms like this
A. Warfarin titrated to an INR goal of 2.0–3.0
before. Her medical history is relevant for hyperlipidemia,
B. Apixaban 2.5 mg orally twice daily
for which she takes atorvastatin 20 mg daily. A 12-lead ECG
C. Catheter ablation
reveals AF with a QRS duration of 90 milliseconds and QTc
D. WATCHMAN device placement
interval of 410 milliseconds. M.E.’s blood pressure is 102/71
mm Hg with heart rate 148 beats/minute. She is resting com- 7. A 62-year-old man (weight 98 kg) has a medical his-
fortably in her hospital bed, and her basic metabolic panel tory that includes hypertension, diabetes, a myocardial
and CBC are within normal limits. infarction (MI) (2 years ago), and paroxysmal AF diag-
nosed 2 months ago. His left ventricular ejection fraction
3. Which one of the following is best to recommend for car-
(LVEF) is 50%. The patient takes losartan 100 mg daily,
dioversion for M.E.?
metformin 1000 mg twice daily, aspirin 81 mg daily, ator-
A. Perform a transesophageal echocardiogram (TEE) vastatin 80 mg daily, metoprolol succinate 200 mg daily,
and sedate for direct current cardioversion (DCCV). and warfarin 7.5 mg once daily (INR 2.7). In the clinic
B. Sedate for DCCV. today, his heart rate is 78 beats/minute, with blood pres-
C. Initiate dronedarone 400 mg orally twice daily. sure 118/80 mm Hg and ECG reflective of rate-controlled
D. Initiate ibutilide 1 mg intravenously over 10 minutes. AF (QRS 110 milliseconds, QTc interval 420 milliseconds).

PSAP 2022 Book 1 • Cardiology 152 Management of Atrial Fibrillation


Despite being rate controlled, the patient continues to 11. A 64-year-old woman is seen in the clinic today in prepa-
have significant episodes of fatigue, dizziness, and palpi- ration for her upcoming AF catheter ablation next week.
tations several times per week and would like to try phar- She takes dabigatran 150 mg orally twice daily. Which
macologic cardioversion. Which one of the following is one of the following is best to recommend to manage
best to recommend for this patient? this patient’s periprocedural anticoagulation?
A. Administer flecainide 100 mg orally twice daily. A. Hold dabigatran 24 hours before AF catheter
B. Administer dronedarone 400 mg orally twice daily. ablation procedure, administer unfractionated
C. Admit to hospital and initiate dofetilide 500 mcg heparin (UFH) to target an activated clotting time
orally twice daily. (ACT) of 300 seconds or more during the procedure,
D. Administer amiodarone 400 mg orally twice daily for then resume dabigatran 3–5 hours after the
2 weeks, then 200 mg once daily. procedure.
B. Change dabigatran to LMWH 12–24 hours before the
Questions 8 and 9 pertain to the following case. procedure and continue LMWH during procedure,
S.N. is a 67-year-old man with hypertension, heart failure then resume dabigatran 3–5 hours after the
with reduced ejection fraction (HFrEF) (LVEF 25%), chronic procedure.
obstructive pulmonary disease, and paroxysmal AF; he pres- C. Continue taking dabigatran uninterrupted, and
ents to the clinic for a follow-up. S.N. takes apixaban 5 mg administer UFH to target an ACT of 300 seconds or
twice daily, metoprolol succinate 200 mg daily, lisinopril greater during the procedure.
40  mg daily, hydrochlorothiazide 25 mg daily, furosemide D. Change dabigatran to warfarin now, and continue
40  mg daily, and ipratropium 2 puffs as needed four times warfarin uninterrupted surrounding ablation
daily. S.N. still has AF-related symptoms several times per procedure, together with UFH to target an ACT of
week, and his vital signs in the clinic today include blood pres- 300 seconds or more during the procedure.
sure 128/78 mm Hg and heart rate 115 beats/minute with an 12. A 58-year-old woman with newly diagnosed paroxysmal
irregularly irregular pulse on auscultation. AF presents to the ED with symptoms of dizziness and
8. Which one of the following is best to recommend for palpitations that began about 1 hour ago. The patient
S.N.? has hypertension and obesity; she takes lisinopril
10  mg daily, apixaban 5 mg twice daily, and metoprolol
A. Digoxin 250 mcg orally once daily
tartrate 25 mg twice daily. Reviewing records of patient
B. Diltiazem 240 mg extended release orally once daily
self-documentation, together with the results of a 7-day
C. Dronedarone 400 mg orally twice daily
Holter monitor, reveals that the patient is symptom-
D. Flecainide 100 mg orally twice daily
atic and aware of when she goes into AF. An ECG today
9. Which one of the following is the optimal target heart reveals AF (ventricular response rate 93 beats/minute)
rate (in beats/minute) to recommend for S.N.? and a QTc of 390 milliseconds, and her vital signs include
A. Less than 60 blood pressure 148/84 mm Hg. In the ED, an oral dose
B. Less than 80 of flecainide 200 mg is administered, which success-
C. Less than 100 fully converts the patient to normal sinus rhythm. The
D. Less than 110 patient wants to try a “pill-in-the-pocket” approach with
flecainide as needed. Which one of the following is best
10. A 57-year-old man is admitted to the hospital for dofeti- to recommend for this patient?
lide initiation. His CrCl is 78 mL/minute, and his baseline
ECG reveals AF (ventricular rate 104 beats/minute) with A. Discontinue metoprolol.
a QTc interval of 430 milliseconds. A repeat ECG checked B. Increase lisinopril to 20 mg daily.
after the first dofetilide dose of 500 mcg reveals a QTc C. Initiate aspirin 81 mg daily.
interval of 505 milliseconds. Which one of the following D. Discontinue apixaban.
is best to recommend for this patient?
A. Continue dofetilide 500 mcg orally twice daily.
B. Decrease dofetilide dose to 250 mcg orally twice
daily.
C. Decrease dofetilide dose to 125 mcg orally twice
daily.
D. Discontinue dofetilide.

PSAP 2022 Book 1 • Cardiology 153 Management of Atrial Fibrillation


13. A 63-year-old man has a history of severe GI bleeding daily (INR 2.3 today), bisoprolol 2.5 mg daily, dronedarone 400
on direct oral anticoagulants (DOACs) (both apixaban mg twice daily, and insulin glargine 30 units daily plus aspart
and  rivaroxaban) and is now being considered for sliding scale with meals. L.D. presents today for her routine
WATCHMAN placement. He currently has normal renal clinic follow-up with no major concerns. Her vital signs and
function and stable vital signs. In considering his ECG reveal blood pressure 121/78 mm Hg and AF (ventricular
post-WATCHMAN placement antithrombotic regimen, rate 100 beats/minute) with a QTc of 425 milliseconds. Infor-
which one of the following is best to recommend for this mation downloaded from her permanent pacemaker shows
patient? that L.D. has been in AF more than 99% of the time for the past
14 months. After discussion with her cardiologist, she does
A. Aspirin 81 mg daily indefinitely plus dabigatran
not want to undergo any more procedures or try any further
150 mg orally twice daily for 45 days, followed by
antiarrhythmic drugs.
clopidogrel 75 mg orally daily for 6 months
B. Aspirin 325 mg orally daily 14. Which one of the following best classifies L.D.’s AF?
C. Aspirin 81 mg daily indefinitely plus clopidogrel
A. Valvular AF
75 mg orally daily for 3 months
B. Longstanding persistent AF
D. Aspirin 81 mg daily indefinitely plus warfarin 5 mg
C. Persistent AF
daily (target INR 2–3) for 45 days, followed by
D. Permanent AF
clopidogrel 75 mg daily for 6 months
15. After this office visit, which one of the following is best to
recommend for L.D.?
Questions 14 and 15 pertain to the following case.

L.D. is an 85-year-old woman whose medical history includes A. Change warfarin to rivaroxaban 15 mg orally daily,
hypertension, AF, diabetes, end-stage renal disease on hemo- with the evening meal.
dialysis, aortic stenosis (moderate), and sick sinus syndrome B. Increase bisoprolol to 5 mg orally daily.
after permanent pacemaker placement 6 years ago. Her C. Discontinue dronedarone.
home drugs include losartan 100 mg daily, warfarin 2.5 mg D. Make no medication adjustments.

PSAP 2022 Book 1 • Cardiology 154 Management of Atrial Fibrillation


Learner Chapter Evaluation: Management of Atrial Fibrillation

As you take the posttest for this chapter, also evaluate the 8. The teaching and learning methods used in the chapter
material’s quality and usefulness, as well as the achievement were effective.
of learning objectives. Rate each item using this 5-point scale: 9. The active learning methods used in the chapter were
effective.
• Strongly agree
10. The learning assessment activities used in the chapter
• Agree
were effective.
• Neutral
• Disagree 11. The chapter was effective overall.
• Strongly disagree 12. The activity met the stated learning objectives.
13. If any objectives were not met, please list them here.
1. The content of the chapter met my educational needs.
2. The content of the chapter satisfied my expectations.
3. The author presented the chapter content effectively.
OTHER COMMENTS
14. Please provide any specific comments related to any
4. The content of the chapter was relevant to my practice
perceptions of bias, promotion, or advertisement of
and presented at the appropriate depth and scope.
commercial products.
5. The content of the chapter was objective and balanced.
15. Please expand on any of your above responses, and/or
6. The content of the chapter is free of bias, promotion, and provide any additional comments regarding this chapter:
advertisement of commercial products.
7. The content of the chapter was useful to me.

PSAP 2022 Book 1 • Cardiology 155 Management of Atrial Fibrillation


Non-Statin Therapy for Dyslipidemia
By Nicholas W. Carris, Pharm.D., BCPS; and Kevin Cowart, Pharm.D., MPH, BCACP, CDCES

Reviewed by John Bucheit, Pharm.D., BCACP, CDCES; Gregory Castelli, Pharm.D., BCPS, BC-ADM; and Eugene N. Bush, Ph.D.,
BCPS, BCCCP

LEARNING OBJECTIVES

1. Distinguish between the drug therapy recommendations of several of the latest and leading guidelines.
2. Justify recommendations for individualized non-statin therapy.
3. Evaluate the role and place in therapy of specific non-statin medications.
4. Develop a comprehensive plan to optimize non-statin therapy.

INTRODUCTION
ABBREVIATIONS IN THIS CHAPTER
Atherosclerotic cardiovascular disease (ASCVD) is the most common
ASCVD Atherosclerotic cardiovascular
disease cause of death in the United States and worldwide. Atherosclerotic
BAS Bile acid sequestrants cardiovascular disease is defined as acute coronary syndrome, myo-
DHA Docosahexaenoic acid cardial infarction, stable or unstable angina, coronary or other arte-
EPA Eicosapentaenoic acid rial revascularization, stroke, transient ischemic attack, or peripheral
FH Familial hypercholesterolemia artery disease, including aortic aneurysm, all of atherosclerotic origin.
Dyslipidemia is one of several factors contributing to the develop-
O3FAs Omega-3 fatty acids
ment and progression of cardiovascular disease. In the United States,
PCSK9 Proprotein convertase subtilisin/
kexin type 9 69.6 million adults have high low-density lipoprotein cholesterol (LDL
RCT Randomized controlled trial cholesterol) (≥ 130 mg/dL), 41.9 million adults have low high-density
lipoprotein cholesterol (HDL cholesterol) (< 40 mg/dL), and 22.2% of
Table of other common abbreviations. adults have high triglycerides (≥ 150 mg/dL) (Virani 2021). Statins are
the primary drug class used for managing dyslipidemia because of
the robust evidence supporting their effectiveness in reducing car-
diovascular events, their low cost, and their convenient daily oral
administration (Grundy 2019). The robust effectiveness for statins in
reducing ASCVD risk is primarily related to their ability to lower LDL
cholesterol. Non-statins can assist in further decreasing LDL choles-
terol to address residual risk. Other non-statin agents are indicated
for patients with elevated triglycerides to reduce both ASCVD and
pancreatic risk.
Before considering non-statin therapy, clinicians must ensure that
statin therapy has been optimized, and, in the case of statin intol-
erance, that several statins have been attempted. Only after these
optimization considerations should non-statin therapy be consid-
ered. Moreover, in cases for which clinicians do consider non-statin
therapies to manage dyslipidemia, the primary goal is prevention
of cardiovascular morbidity and mortality whereas the second-
ary goal is prevention of pancreatitis. Therefore, non-statin ther-
apy is reserved for patients who meet one or more of the following
criteria: 1) insufficient response to statin therapy; 2) intolerance to
statin therapy; 3)  severe hypertriglyceridemia, and 4) indicated use

PSAP 2022 Book 1 • Cardiology 157 Non-Statin Therapy for Dyslipidemia


of icosapent ethyl to lower cardiovascular risk. In all cases their LDL cholesterol threshold. Second, not all patients will
of management with non-statin therapy, agents with cardio- achieve the expected LDL cholesterol reduction based on
vascular benefit established in randomized controlled trials statin regimen potency, even with good adherence to ther-
(RCTs) should be used preferentially. apy. Thus, patients with residually elevated LDL cholesterol
(e.g., ≥ 70 mg/dL in a patient with ASCVD) may have the opportu-
POPULATIONS LIKELY TO BENEFIT nity to further reduce their cardiovascular risk with additional
FROM NON-STATIN THERAPY therapy. Although medication-directed versus LDL cholesterol-
A pathway summarizing the assessments and non-statin directed approaches to care have been debated, a growing
treatment options is presented in Figure 1. body of literature supporting very low LDL cholesterol as ben-
eficial was recently summarized (Karagiannis 2021). Finally,
Insufficient Response to Statin Therapy some patients may not be able to tolerate the statin intensity
indicated for their disease and thus are more likely to have
Insufficient responses to statin therapy can be grouped
an LDL cholesterol that remains above the desired threshold.
into three categories, although each are addressed simi-
Across recent guidelines (Table 1 and Table 2), the most
larly by the addition of non-statin therapy. First, patients
common goal LDL cholesterol concentrations can be classi-
may have a high baseline LDL cholesterol, such that even
fied into two groups as follows: 1) in the primary prevention of
with a 50% reduction from baseline in LDL cholesterol with a
ASCVD, an LDL cholesterol goal less than 100 mg/dL, and 2) in
high-intensity statin, LDL cholesterol remains above a desired
the secondary prevention of ASCVD, an LDL cholesterol goal
threshold. Patients with homozygous or heterozygous famil-
less than 70 mg/dL. To interpret these goals, knowledge of
ial hypercholesterolemia (FH) are likely to need non-sta-
risk factors is needed and is most conveniently summarized
tin therapy in addition to an optimized statin to surpass
by factors include in the ASCVD Risk Estimator Plus and “risk
enhancing factors” listed and described in the 2018 AHA/ACC
Guideline on the Management of Blood Cholesterol.
BASELINE KNOWLEDGE STATEMENTS However, LDL cholesterol goals may be better viewed as
“thresholds” that, if not achieved, warrant consideration of
Readers of this chapter are presumed to be familiar
with the following: the addition of non-statin therapy. This approach is in con-
trast to a typical “goal” because some patients may have rela-
• Fundamental knowledge of lipid-lowering medica- tively low LDL cholesterol at baseline, yet initial statin therapy
tions, their lipid-lowering effects, their safety
profile, and guideline-directed use of older is based on cardiovascular risk; therefore, initial therapy may
lipid-lowering medications, such as fibrates often be the same as for a patient with relatively higher base-
• General treatment strategies, cardiovascular risk line LDL cholesterol. For example, a statin-naive patient
assessment, and cardiovascular risk groups estab- status post-myocardial infarction with a baseline LDL choles-
lished in the 2018 ACC/AHA Blood Cholesterol terol of 99 mg/dL may be able to achieve an LDL cholesterol
Guideline less than 70 mg/dL with a moderate-intensity statin; however,
• Pathophysiology of atherosclerotic cardiovascular based on the patient’s cardiovascular risk, a high-intensity
disease
statin is indicated. The “goal” of LDL cholesterol of less
Table of common laboratory reference values than 70 mg/dL only becomes relevant if the optimized statin
dose (in this case, a high-intensity statin) does not achieve
an LDL cholesterol of less than 70 mg/dL. The potential to
ADDITIONAL READINGS achieve a goal LDL cholesterol with a lower potency statin
than indicated based on cardiovascular risk, in either pri-
The following free resources have additional back-
ground information on this topic: mary or secondary prevention, should not be a rationale to
underdose statin therapy. Similarly, the potential to achieve
• 2018 AHA/ACC Guideline on the Management of
Blood Cholesterol. an LDL cholesterol goal with an intentionally underdosed
statin plus non-statin therapy is not warranted. Conversely, in
• Cardiovascular Disease and Risk Management:
Standards of Medical Care in Diabetes—2021. a patient with ASCVD who is unable to tolerate high-intensity
• 2013 AHA/ACC/TOS Guideline for the Management statin therapy, it is reasonable to treat with the maximum
of Overweight and Obesity in Adults. tolerated statin (e.g., moderate-intensity statin), then add
• 2017 Focused Update of the 2016 ACC Expert Con- non-statin therapy to achieve the recommended LDL cho-
sensus Decision Pathway on the Role of Non-Statin lesterol reduction. These thresholds and approaches closely
Therapies for LDL-Cholesterol Lowering in the Man- relate between the American Diabetes Association standards
agement of Atherosclerotic Cardiovascular Disease of care and the multi-society blood cholesterol guidelines.
Risk.
However, the American Association of Clinical Endocrinolo-
gists and American College of Endocrinology joint consensus

PSAP 2022 Book 1 • Cardiology 158 Non-Statin Therapy for Dyslipidemia


A.

Assess ASCVD Risk, Baseline and Current Lipid


Panel, Current and Past Lipid Therapies, Current
Statin Intensity, and Indicated Statin Intensity

Is Patient on Indicated Statin Therapy or Yes


Maximally Tolerated Statin Therapy AND is their
LDL-C at Goal Based on ASCVD Risk?

No
Maximize Statin Therapy

Does the Patient Need a 25% Reduction in LDL-C Evaluate for


or More to Reach Their Goal? Residual
ASCVD
Prefer Prefer Risk
In In Reduction
No Primary Secondary Yes and
Prevention Prevention Triglyceride
And FH Targeted
Therapy
Consider Ezetimibe, Consider a PCSK9 (Box 3B)
Alternatively or Inhibitor, Alternatively
Subsequently a PCSK9 or Subsequently
Inhibitor Can be Used Ezetimibe Can be Used

If Not at Goal, Consider Bile Acid Sequestrant,


Bempedoic Acid, or Referral to Lipid Specialist

Figure 1. Pathways for implementing non-statin therapy: A,  LDL cholesterol lowering with non-statin therapy;
B, residual ASCVD risk reduction and triglyceride targeted therapy.
Information from: Bays HE, Braeckman RA, Ballantyne CM, et al. Icosapent ethyl, a pure EPA omega-3 fatty acid: effects on
lipoprotein particle concentration and size in patients with very high triglyceride levels (the MARINE study). J Clin Lipidol
2012;6:565-72; Bhatt DL, Steg PG, Miller M, et al. Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N Engl
J Med 2019;380:11-22; Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APHA/ASPC/
NLA/PCNA guideline on the management of blood cholesterol: a report of the American College of Cardiology/American Heart
Association Task Force on clinical practice guidelines. Circulation 2019;139:e1082-143; Handelsman Y, Jellinger PS, Guerin CK, et al.
Consensus statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the
management of dyslipidemia and prevention of cardiovascular disease algorithm—2020 executive summary. Endocr Pract
2020;26:1196-224; Harris WS, Ginsberg HN, Arunakul N, et al. Safety and efficacy of Omacor in severe hypertriglyceridemia. J
Cardiovasc Risk 1997;4:385-91; Jacobson TA, Ito MK, Maki KC, et al. National Lipid Association recommendations for patient-
centered management of dyslipidemia: part 1—full report. J Clin Lipidol 2015a;9:129-69; Jacobson TA, Maki KC, Orringer CE, et al.
National Lipid Association recommendations for patient-centered management of dyslipidemia: part 2. J Clin Lipidol 2015b;9:S1-
122.e1.

(continued)

statement offers an even lower threshold (LDL cholesterol < (PCSK9) inhibitors evolocumab and alirocumab (Schwartz
55 mg/dL) for patients at the highest cardiovascular risk. 2018; Sabatine 2017). Alternatively, ezetimibe is an option
After confirming that statin therapy has been optimized, with evidence of cardiovascular benefit, although it offers
non-statin therapy can be considered. The next class of less LDL cholesterol reduction compared with PCSK9 inhib-
agents with the most compelling evidence of cardiovascular itors (Handelsman 2020; Cannon 2015). For patients near
benefit are the proprotein convertase subtilisin/kexin type 9 their LDL cholesterol threshold, ezetimibe may be preferred

PSAP 2022 Book 1 • Cardiology 159 Non-Statin Therapy for Dyslipidemia


B.

Is the Patient Indicated for Icosapent Ethyl for ASCVD Risk Reduction? Atherosclerotic cardiovascular
disease, ASCVD
- Triglycerides 135–499 mg/dL, and
- Maximally tolerated statin, and Docosahexaenoic acid, DHA
- ASCVD or Diabetes plus at least 2 ASCVD risk factors Eicosapentaenoic acid (EPA)

No Yes Low density lipoprotein


cholesterol, LDL-C
Does the Patient have Start Icosapent Ethyl for Proprotein convertase
Serum Triglycerides ASCVD Risk Reduction subtilisin/kexin type 9, PCSK9
500 mg/dL or greater?

Yes No

Does the Patient have


Continue to Monitor Yes Implement Triglyceride Targeted Lifestyle
Response to Serum Triglycerides Intervention, Consider Fibrate Therapy
Therapy, Implement 500 mg/dL or greater? and Referral to Lipid Specialist
Lifestyle
Intervention No

Continue to Monitor Response to Therapy, Implement Lifestyle Intervention

Does the Patient have


Serum Triglycerides Yes Start Prescription Combination EPA/DHA Product and Implement
650 mg/dL or greater? Triglyceride Targeted Lifestyle Intervention
No

Start lcosapent Ethyl for Does the Patient have Yes Consider Fibrate Therapy and
Triglyceride Reduction and Serum Triglycerides Referral to Lipid Specialist,
Implement Triglyceride Targeted 500 mg/dL or greater? Continue to Monitor Response to
Lifestyle Intervention Therapy
No

Continue to Monitor
Response to Therapy

Figure 1. Pathways for implementing non-statin therapy: A,  LDL cholesterol lowering with non-statin therapy;
B, residual ASCVD risk reduction and triglyceride targeted therapy.
Information from: Bays HE, Braeckman RA, Ballantyne CM, et al. Icosapent ethyl, a pure EPA omega-3 fatty acid: effects on
lipoprotein particle concentration and size in patients with very high triglyceride levels (the MARINE study). J Clin Lipidol
2012;6:565-72; Bhatt DL, Steg PG, Miller M, et al. Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N Engl
J Med 2019;380:11-22; Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APHA/ASPC/
NLA/PCNA guideline on the management of blood cholesterol: a report of the American College of Cardiology/American Heart
Association Task Force on clinical practice guidelines. Circulation 2019;139:e1082-143; Handelsman Y, Jellinger PS, Guerin CK, et al.
Consensus statement by the American Association of Clinical Endocrinologists and American College of Endocrinology on the
management of dyslipidemia and prevention of cardiovascular disease algorithm—2020 executive summary. Endocr Pract
2020;26:1196-224; Harris WS, Ginsberg HN, Arunakul N, et al. Safety and efficacy of Omacor in severe hypertriglyceridemia. J
Cardiovasc Risk 1997;4:385-91; Jacobson TA, Ito MK, Maki KC, et al. National Lipid Association recommendations for patient-
centered management of dyslipidemia: part 1—full report. J Clin Lipidol 2015a;9:129-69; Jacobson TA, Maki KC, Orringer CE, et al.
National Lipid Association recommendations for patient-centered management of dyslipidemia: part 2. J Clin Lipidol 2015b;9:S1-
122.e1.

over PCSK9 inhibitors because of its lower cost and the con- Statin Intolerance
venience of oral administration. Of note, patients treated with A leading cause for statin intolerance is muscle symptoms,
PCSK9 inhibitors may achieve very low LDL cholesterol con- primarily myalgia, which can reduce statin adherence and
centrations. Concentrations as low as 20 mg/dL are consid- worsen cardiovascular outcomes (Box 1). In practice, up to
ered safe and beneficial, although with some potential risks 30% of patients experiences challenges in tolerating statin
(Karagiannis 2021; Giugliano 2017b). therapy; however, this difficulty may be attributed to many

PSAP 2022 Book 1 • Cardiology 160 Non-Statin Therapy for Dyslipidemia


Table 1. Summary of Guideline Recommendations for Managing Blood LDL Cholesterol for the Primary Prevention of
Atherosclerotic Cardiovascular Disease

Guideline Primary Prevention of ASCVD

2021 ADA: With diabetes: With diabetes: With diabetes: Adults with diabetes; 10-year
Standards of Age 20–39 yr; ASCVD Age 40–75 yr; use At higher ASCVD risk; ASCVD risk 20% or higher;
Medical Care in risk factors; moderate-intensity High-intensity statin May be reasonable to add
Diabetes Statin therapy may be statin therapy is reasonable ezetimibe to maximally
reasonable tolerated statin therapy
to reduce LDL-C by 50% or
more

2020 AACE/ACE: No risk factors: < 2 risk factors and 2 or more risk factors Diabetes with at least 1
Management of LDL-C Goal is < 130 10-year risk < 10%; and 10-year risk risk factor OR CKD at least
Dyslipidemia and mg/dL LDL-C Goal is < 100 10-20% OR diabetes stage 3 with albuminuria OR
Prevention of mg/dL OR CKD; heterozygous FH OR ASCVD
Cardiovascular LDL-C Goal < 100 mg/dL risk > 20%:
Disease LDL-C Goal < 70 mg/dL

To reach goal: Moderate-Intensity Statin→ High-Intensity Statin→ add PCSK9i→ add ezetimibea

2018 AHA/ACC/ Without Diabetes or Without diabetes or With diabetes; without LDL-C of 190 mg/dL or more;
AACVPR/AAPA/ LDL-C of 190 mg/dL LDL-C of 190 mg/dL LDL-C of 190 mg/dL high-intensity statin; add
ABC/ACPM/ or greater; 10-year or greater; 10-year or greater; moderate- ezetimibe if LDL-C not
ADA/AGS/ ASCVD risk of 5% ASCVD risk of 20% or intensity statin; reduced 50% or if LDL-C
APhA/ASPC/ to < 20%; assess more; high-intensity Consider high-intensity remains > 100 mg/dL;
NLA/PCNA cardiovascular risk statin; if would benefit statin based on Triple therapy options:
Guideline on the factors not accounted from high-intensity ASCVD risk; if high- a) if LDL-C still not reduced
Management for in risk calculator; statin, but cannot risk, can consider 50% and TGs 300 mg/dL or
of Blood consider moderate- tolerate, can consider adding ezetimibe to less consider BAS
Cholesterol intensity statin adding ezetimibe achieve 50% reduction b) if heterozygous FH and
in LDL-C LDL-C 100 mg/dL or
greater, consider PCSK9i
Age 40–75 yr
c) if baseline LDL-C was
220 mg/dL or greater and
LDL-C remains 130 mg/dL
or greater consider PCSK9i

a
Ezetimibe may be preferred as second-line therapy because of low cost in patients needing 20% LDL cholesterol reduction or less.
ASCVD = atherosclerotic cardiovascular disease; BAS = bile acid sequestrant; CKD = chronic kidney sisease; FH = familial
hypercholesterolemia; LDL-C = LDL cholesterol; PCSK9i = proprotein convertase subtilisin/kexin type 9 inhibitor; TGs = triglycerides.
Information from: American Diabetes Association. Cardiovascular disease and risk management: standards of medical care in
diabetes-2021. Diabetes Care 2021;44(Suppl 1):S125-S150; Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/
ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: a report of the American College
of Cardiology/American Heart Association task force on clinical practice guidelines. Circulation. 2019;139:e1082-e1143;
Handelsman Y, Jellinger PS, Guerin CK, et al. Consensus statement by the American Association of Clinical Endocrinologists and
American College of Endocrinology on the management of dyslipidemia and prevention of cardiovascular disease algorithm - 2020
executive summary. Endocr Pract 2020;26:1196-2.

(continued)
causes, including the nocebo effect, based on the observa- a “nocebo” effect (Herrett 2021; Wood 2020). The nocebo
tion that only about 10% of patients in RCTs report statin intol- effect may arise with statin therapy because of medication
erance (Newman 2019). In brief, the nocebo effect entails the counseling provided to the patient regarding potential muscle
patient expectation of myalgia that subsequently results in effects, the patient’s exposure to negative media regarding
the actual experience of myalgia. Two recent major publica- statins and muscle effects, or the patient’s discussion with
tions suggest that many statin-treated patients who report peers regarding statins and muscle effects.
myalgia may be experiencing these muscle symptoms not Two trials recently tested the nocebo effect using a method
because of the effects of the statin, but instead because of known as a series of “n-of-1” trials (Herrett 2021; Wood 2020).

PSAP 2022 Book 1 • Cardiology 161 Non-Statin Therapy for Dyslipidemia


Table 2. Summary of Guideline Recommendations for Managing Blood LDL Cholesterol for the Secondary Prevention of
Atherosclerotic Cardiovascular Disease

Guideline Secondary Prevention of ASCVD

2021 ADA: Standards With diabetes; high-intensity With diabetes; if very high risk with ASCVD; if LDL-C is 70 or greater
of Medical Care in statin for all patients on maximally tolerated statin consider adding ezetimibe or
Diabetes PCSK9i; ezetimibe may be preferred because of lower cost

2020 AACE/ACE: Recent hospitalization for LDL-C goal is < 55 mg/dL in three cases
Management of acute coronary syndrome OR a. Progressive ASCVD;
Dyslipidemia and established clinical ASCVD; b. E
 stablished clinical ASCVD AND diabetes OR CKD stage 3 or
Prevention of LDL-C goal is < 70 mg/dL greater OR heterozygous FH;
Cardiovascular Disease c. P
 remature ASCVD age < 55 yr (men), age < 65 yr (women);

To reach goal: High-Intensity Statin→ add PCSK9i→ add ezetimibea,b

2018 AHA/ACC/AACVPR/ ASCVDc and age over 75 yr; ASCVD and age 75 yr and Very high risk ASCVD; high-
AAPA/ABC/ACPM/ADA/ start moderate-or high- less; high-intensity statin or intensity statin or maximum
AGS/APhA/ASPC/NLA/ intensity statin; continue maximum tolerated statin; tolerated statin; if LDL-C remains
PCNA Guideline on the high-intensity statin if LDL-C remains > 70 mg/dL > 70 mg/dL consider adding
Management of Blood consider adding ezetimibe ezetimibe; if LDL-C remains > 70
Cholesterol mg/dL consider added PCSK9i

a
Ezetimibe may be preferred as second-line therapy because of low cost in patients needing 20% LDL-C reduction or less
b
Consider colesevelam or bempedoic acid as alternative third-line agent to ezetimibe or PCSK9 inhibitor if needed
c
With atherosclerotic origin, includes the following conditions: acute coronary syndrome, myocardial infarction, stable or unstable
angina, coronary or other arterial revascularization, stroke, transient ischemic attack, peripheral artery disease, and aortic aneurysm.
ASCVD = atherosclerotic cardiovascular disease; CKD = chronic kidney disease; FH = familial hypercholesterolemia; LDL-C = LDL
cholesterol; PCSK9i = proprotein convertase subtilisin/kexin type 9 inhibitor.
Information from: American Diabetes Association. 10. Cardiovascular disease and risk management: standards of medical care in
diabetes-2021. Diabetes Care. 2021;44(Suppl 1):S125-S150; Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/
ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: a report of the American College
of Cardiology/American Heart Association task force on clinical practice guidelines. Circulation 2019;139(25):e1082-e1143;
Handelsman Y, Jellinger PS, Guerin CK, et al. Consensus statement by the American Association of Clinical Endocrinologists and
American College of Endocrinology on the management of dyslipidemia and prevention of cardiovascular disease algorithm—2020
executive summary. Endocr Pract 2020;26:1196-224.

Box 1. Definitions of SAMSa


2002 ACC/AHA Advisory on Statins • Myopathy: Weakness
• Myopathy: Any muscle symptoms (SAMS) • Myositis: Inflammation
• Myalgia: SAMS CK = NL • Myonecrosis: CK 3 × ULN
• Myositis: SAMS CK > ULN ○ Mild: CK > 3, < 10 × ULN
• Rhabdomyolysis: CK > 10 × ULN ○ Moderate: CK > 10, < 50 × ULN
○ Severe: CK > 50 × ULN
2013 Canadian Work Group
○ Rhabdomyolysis: CK > ULN and SCr > 0.5 mg/dL baseline
• Myopathy: Any muscle symptoms (SAMS)
• Symptomatic myalgia: 2018 ACC/AHA Cholesterol Guidelines
○ Myalgia CK ≤ ULN • Myalgias: CK = NL
○ Myositis CK > ULN • Myositis/myopathy: CK > ULN with symptoms of concern or
○ Rhabdomyolysis CK > 10 × ULN objective weakness

2014 NLA Muscle Safety Task Force • Rhabdomyolysis: CK >10 × ULN + renal injury
• Myalgia: Aching, stiffness, cramps
a
Normal CK range is 38–174 units/L for men and 96–140 units/L for women.
ACC = American College of Cardiology; AHA = American Heart Association; SAMS = statin-associated muscle symptoms; NL = normal
limit; NLA = National Lipid Association; ULN = upper limit of normal.
Reprinted from: Beavers CJ, Kelly MS. Dyslipidemia. In: Murphy JE, Lee MW, eds. Pharmacotherapy Self-Assessment Program, 2019 Book
1. Cardiology. Lenexa, KS: American College of Clinical Pharmacy, 2019:31-54.

PSAP 2022 Book 1 • Cardiology 162 Non-Statin Therapy for Dyslipidemia


The trials included patients who had stopped or were consid- not support their use, including vitamin D or coenzyme Q10
ering stopping statin therapy because of adverse effects. In (Grundy 2019). In cases of severe muscle symptoms, cre-
these studies, patients received blinded treatment with ator- atinine kinase values should be measured. Notably, statins
vastatin 20 mg tablets and placebo, alternating through sev- are associated with more severe muscle conditions, such as
eral phases. One of the trials also included a no-treatment myopathy, myositis, rhabdomyolysis, and statin-associated
phase. While rotating through randomized, blinded phases, autoimmune myopathy, all of which are possible, but rare,
patients reported their muscle symptoms. In both trials, patient with each requiring their own management (Newman 2019).
reported muscle symptoms were similar during the placebo
and statin phase, suggesting that statin-related myalgia may Non-Statin Recommendations
be the result of a nocebo effect. However, the trials were not for Hypertriglyceridemia
without weakness. Although power is greatly increased by the Apart from cardiovascular risk reduction, non-statin thera-
multiple paired comparison, with each patient serving as their pies can be selectively used in the management of very high
own control for several comparisons, one trial included only triglycerides (≥ 1000 mg/dL) to reduce the risk for pancre-
60 patients. The second trial was only able to analyze data for atitis (Jacobson 2015a). Serum triglyceride concentrations
151 of 200 intended patients; only 114 patients completed all of 1000 mg/dL or greater should be managed with prescrip-
six treatment periods. Among the 114 patients, 17 had average tion omega-3-acid ethyl esters (O3FAs), icosapent ethyl, or
muscle symptoms at least one unit higher during statin ther- fibrates. For patients with serum triglyceride concentrations
apy versus placebo, suggesting that statins may be contrib- 500–999 mg/dL and an ASCVD risk of 7.5% or higher, statin
uting to muscle symptoms for these patients (Herrett 2021). therapy is often sufficient for cardiovascular risk reduction,
Another weakness is that the studies used atorvastatin although high-intensity statin therapy may be needed for
20 mg/day. Because of the potential for the adverse effects sufficient triglyceride reduction. Non-statin therapy may be
of a statin on muscles to be dose related and because the required for additional lowering of triglycerides to achieve
reports of muscle pain overall were low in both trials, it is pos- a concentration less than 500 mg/dL after attempting to
sible that the results may have differed with a higher ator- address the underlying cause(s) for a patient’s high tri-
vastatin dose. Despite these weaknesses, on unblinding and glycerides, including therapeutic lifestyle changes. Patients
counseling, 34 patients (57% of all patients) in one trial and 65 whose triglycerides remain high may have success with ico-
patients (68% of patients informed that statins were unlikely spent ethyl to lower triglycerides (as discussed in the follow-
to be contributing to their muscle symptoms) in the other trial ing section). Or, in the case of very high triglycerides, patients
had restarted or planned to restart statin therapy. Therefore, may benefit more with respect to triglyceride reduction and
it is possible that if “n-of-1 packs” were made available, clini- the decreased risk of pancreatitis from treatment with typi-
cians could use this approach to assess muscle symptoms cal prescription O3FA products containing eicosapentaenoic
and counsel patients to increase statin use. After attempts acid (EPA) and docosahexaenoic acid (DHA) because tri-
to optimize statin therapy, non-statin options are the same as glycerides can be reduced by as much as 45% depending on
those for an insufficient statin response. baseline triglyceride concentration (Harris 1997). Fibrates are
In context on these considerations, clinicians should alternative agents that include fenofibrate, which can be used
obtain a detailed history of muscle symptoms at baseline, with statins, and gemfibrozil, which should not be used with
including current muscle symptoms and activities known to statins. For patients with triglyceride concentrations less
provoke muscle symptoms, to avoid misattribution of myal- than 500 mg/dL, cardiovascular risk reduction is the sole con-
gia to statin therapy. The baseline history should be paired cern, and statin therapy based on cardiovascular risk should
with detailed counseling on the risks and benefits of statin predominate. For patients with triglycerides concentrations
therapy to prevent undue statin interruption or discontinua- less than 500 mg/dL, therapeutic lifestyle changes are the
tion. Statins are more likely the cause of myalgia if the pain primary intervention to directly address high triglycerides.
is bilateral, involves proximal muscles, has an onset within
weeks or months of statin therapy, and improves on statin Indication Directed Use of Icosapent Ethyl
discontinuation. Neither the potential for adverse effects The REDUCE-IT trial (discussed in detail in the Omega-3 Fatty
of statins nor the adverse effects associated with one sta- Acids section) demonstrated the cardiovascular benefit of
tin should typically dissuade clinicians from pursuing sta- icosapent ethyl (Bhatt 2019). Based on these data, icosapent
tin therapy (Grundy 2019). Indeed, a trial of as many different ethyl has been incorporated into several society guidelines
statins as a patient is willing to attempt is often reasonable for ASCVD risk reduction, although for specific patients who
in response to non-severe adverse effects before concluding match the REDUCE-IT trial inclusion and exclusion criteria
that a patient has complete statin intolerance (Nissen 2016). (American Diabetes Association 2021b; Handelsman 2020).
For the management of non-severe muscle symptoms, sev- However, in considering the tandem goal of lowering tri-
eral approaches have been attempted in addition to trials of glycerides to less than 500 mg/dL and to decrease both the
several different statins. However, current data largely do pancreatitis risk and ASCVD risk, several considerations are

PSAP 2022 Book 1 • Cardiology 163 Non-Statin Therapy for Dyslipidemia


key. Notably, the multi-society blood cholesterol guideline
Box 2. Non-Statin Drugs with Evidence
was published before the finding that icosapent ethyl low-
Supporting ASCVD Risk Reductiona
ered the cardiovascular risk; therefore, for pharmacologic
• PCSK9 inhibitors management of triglycerides, the guideline focused on low-
• Ezetimibe ering triglycerides less than 500 mg/dL to reduce pancreati-
• Icosapent ethyl
• Bile acid sequestrants tis risk (Bhatt 2019; Grundy 2019). In addition, because of
• Gemfibrozil the potential for cardiovascular benefit, icosapent ethyl may
be preferred in some circumstances for triglyceride lower-
a
Listed in order of preference. ing, despite no reflection of this preference in the multi-soci-
ASCVD = atherosclerotic cardiovascular disease; PCSK9 =
proprotein convertase subtilisin/kexin type 9.
ety guideline. However, use of icosapent ethyl in populations

Table 3. Non-Statin Pharmacotherapy

Drug Class Dosing Impact on Lipids Considerations

PCSK9 Inhibitors

Alirocumab • Initial: 75 mg SQ every • Mean LDL-C • Adverse effects


2 wk; may increase to reduction up to 60% ○ Alirocumab: nasopharyngitis, injection site

150 mg every 2 wk • Mean TG reduction reactions, influenza


• Alternative initial dose is up to 17% ○ Evolocumab: nasopharyngitis, upper

300 mg SQ every 4 wk • Mean non-HDL-C respiratory tract infection, influenza, back


reduction up to 53% pain, and injection site reactions
Evolocumab • ASCVD or Heterozygous
• No clinically significant drug–drug
FH: 140 mg SQ every 2 wk
interactions
or 420 mg SQ once monthly
• Other considerations: cost/prior authorization
• Homozygous FH: 420 mg
burden, injectable therapy
SQ once monthly

Omega-3 Fatty Acids

Omega-3-ethyl 4 g/day • Mean TG reduction • Adverse effects


esters (Lovaza) up to 50% ○ Lovaza: burping, dyspepsia, fishy aftertaste

• Mean HDL-C ○ Vascepa: musculoskeletal pain, peripheral

increase up to 9% edema, constipation, gout


• May increase LDL-C • Drug interactions: antiplatelets (increased
bleeding risk)
• Other considerations: may increase atrial
Icosapent Ethyl 4 g/day • Mean TG reduction
fibrillation risk (Lovaza and Vascepa); use
(Vascepa) up to 27%
caution with fish/shellfish allergy
• May decrease HDL-C
and increase LDL-C

Cholesterol Absorption Inhibitor

Ezetimibe 10 mg/day with or without • Mean LDL-C • Adverse effects


food reduction up to 20% ○ Monotherapy: upper respiratory tract

monotherapy (25% infection, diarrhea, arthralgia, sinusitis,


when used with a pain in extremity
statin) ○ In combination with statin:

• Mean HDL-C nasopharyngitis, myalgia, upper respiratory


increase up to 4% tract infection, arthralgia, diarrhea
• Mean TG reduction • Drug–drug interactions: cyclosporine,
up to 10% fibrates, cholestyramine (separate by ≥ 2 hr
before or ≥ 4 hr after BAS dose)
• Other considerations: well tolerated, available
as generic; contraindicated in active liver
disease, and pregnancy/breastfeeding

(continued)

PSAP 2022 Book 1 • Cardiology 164 Non-Statin Therapy for Dyslipidemia


Table 3. Non-Statin Pharmacotherapy  (continued)

Drug Class Dosing Impact on Lipids Considerations

ACL Inhibitor

Bempedoic acid 180 mg/day with or without • Mean LDL-C • Adverse effects: gout, tendon rupture, upper
food reduction up to 23% respiratory tract infection, abdominal/back
(monotherapy), pain, anemia
up to 48% (with • Drug–drug interactions: statins
ezetimibe), up to • Considerations: pregnancy and breastfeeding
27% (with PCSK9
inhibitor), and up to
24% (with statin)
• Mean non-HDL-C
reduction up to 12%

BAS

Cholestyramine 8–16 g/day divided into • Mean LDL-C • Adverse effects: constipation, dyspepsia,
2 doses reduction up to 30% abdominal pain, and nausea
• Mean HDL-C • Drug–drug interactions: phenytoin, warfarin,
Colestipol 2–16 g/day given once or in
increase up to 5% digoxin, levothyroxine, fat-soluble vitamins
divided doses
• May increase TG (A, D, E, K)
Colesevelam • Tablets: 6 tablets daily or • Drugs with potential interaction should be
3 tablets twice daily; take taken ≥ 1 hr before or ≥ 4 hr after BAS to avoid
tablets with a meal and impeding their absorption
liquid • Other considerations:
• Suspension: one 3.75-g ○ Inconvenient preparation: pill burden with

packet daily, or one tablets and oral suspension


1.875-g packet twice daily; ○ Colesevelam lowers A1C by about 0.5% and

mix powder with 4–8 oz has fewer GI adverse effects than other
of water, fruit juice, or soft BAS
drink; take with meal ○ Cholestyramine is contraindicated with

• Note: 3.75 g is equivalent compete biliary obstruction


to 6 tablets, and 1.875 g is ○ Colesevelam is contraindicated with bowel

equivalent to 3 tablets obstruction

Fibrates

Gemfibrozil 600 mg twice daily • Mean HDL-C • Adverse effects: mild GI, dyspepsia, myopathy
increase up to 15% • Drug–drug interactions
Fenofibrate 48–145 mg once daily
• Mean TG decrease ○ Warfarin, immunosuppressants, BAS

up to 50% ○ Gemfibrozil: avoid with lovastatin,

Fenofibric acid 35–105 mg once daily pravastatin, simvastatin; may be


acceptable with atorvastatin, rosuvastatin,
and pitavastatin if fibrate needed and
fenofibrate is not an option
• Other considerations: contraindicated
in breastfeeding, severe liver, renal and
gallbladder disease; may increase liver
transaminases

ACL = adenosine triphosphate-citrate lyase; ASCVD = atherosclerotic cardiovascular disease; BAS = bile acid sequestrants;
FH = familial hypercholesterolemia; HDL-C = high-density lipoprotein cholesterol; LDL-C = LDL cholesterol; PCSK9 = proprotein
convertase subtilisin/kexin type 9; SQ = subcutaneous; TG = triglycerides.
Information from: Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APHA/ASPC/NLA/
PCNA guideline on the management of blood cholesterol: a report of the American College of Cardiology/American Heart
Association Task Force on clinical practice guidelines. Circulation 2019;139:e1082-143; manufacturers’ package inserts.

PSAP 2022 Book 1 • Cardiology 165 Non-Statin Therapy for Dyslipidemia


beyond the REDUCE-IT trial inclusion/exclusion criteria may unstable angina, or coronary revascularization. Evolocumab
or may not experience the same cardiovascular benefit. significantly reduced the risk of the composite primary end
point (absolute risk reduction [ARR] 1.5%; HR 0.85; 95% CI,
0.79–0.92) and reduced the composite of cardiovascular
NON-STATIN AGENTS death, myocardial infarction, or stroke (ARR 1.5%; HR 0.80;
FOR DYSLIPIDEMIA 95% CI, 0.73–0.88). Although adverse events did not differ by
In choosing non-statin agents for the treatment of dyslipid- subgroups stratified by minimum LDL cholesterol achieved,
emia, evidence supporting the reduction of ASCVD risk is the concerns were raised over the low LDL cholesterol reached
primary deciding factor (Box 2). In Figure 1 and Table 3, the evi- in FOURIER and the impact on cognition. The median LDL
dence is summarized to facilitate this decision-making pro- cholesterol attained in the evolocumab arm was 30 mg/dL;
cess in a patient-specific manner. In addition, older agents, however, the EBBINGHAUS study, which analyzed a sub-
some options and some to be avoided, are addressed. As a group of patients from the FOURIER trial, found no significant
part of choosing non-statin medications to reduce ASCVD between-group difference in cognitive function over a median
risk, the benefit of some agents may be limited to patients of 19 months (Giugliano 2017a). Evolocumab has also been
with specific dyslipidemia patterns. Finally, safety and drug shown to reduce the LDL cholesterol with a similar adverse
interactions must also be taken into account. For patients event profile to placebo in pediatric patients with FH.
with very high triglyceride concentrations, specific pharma- The PCSK9 inhibitors, which are available as subcutane-
cologic treatment to reduce the risk of pancreatitis should be ous injections and administered every 2–4 weeks, are gener-
considered. ally well tolerated. The most common adverse drug reactions
are injection site reactions, immunologic or allergic reac-
PCSK9 Inhibitors tions, and nasopharyngitis, according to the manufacturers’
The LDL receptors on hepatocytes are degraded by the enzyme package inserts. Injection site reaction event rates in the
PCSK9, which is predominately produced in the liver. By FOURIER and ODYSSEY trials for treatment versus placebo
inhibiting the PCSK9 enzyme, more LDL receptors are avail- were 2.1% vs. 1.6% and 3.8% vs. 2.1%, respectively (Schwartz
able to remove plasma LDL cholesterol, as described in the 2018; Sabatine 2017). Monoclonal antibodies are not elimi-
manufacturers’ package inserts. When combined with sta- nated by the kidneys and can therefore be used in patients
tin therapy, PCSK9 inhibitors provide up to an additional 60% with mild to moderate renal impairment, although they are
reduction in LDL cholesterol and can lower triglycerides by up not well studied in patients with severe renal impairment,
to 17%. Currently available PCSK9 inhibitors are alirocumab according to the manufacturers’ package inserts. A major
and evolocumab, which are humanized monoclonal antibodies. limitation to using PCSK9 inhibitors is their high cost. The
The ASCVD benefit of alirocumab was demonstrated in the cost-effectiveness of PCSK9 inhibitors for primary preven-
ODYSSEY OUTCOMES trial (Schwartz 2018). Patients with an tion among patients with an LDL cholesterol greater than
acute coronary syndrome in the previous 12 months with an 190 mg/dL or in those with FH has not been evaluated exten-
LDL cholesterol 70 mg/dL or greater, or a non-HDL cholesterol sively. However, because the cost of PCSK9 inhibitors has
100 mg/dL or greater, or apolipoprotein B 80 mg/dL or greater decreased since the publication of the 2018 American Heart
were randomly assigned to receive alirocumab added to their Association/American College of Cardiology guidelines,
current maximally tolerated statin and ezetimibe therapy. The cost-effectiveness is now acceptable in those with very
composite primary end point was death from coronary heart high-risk ASCVD (Fonarow 2019).
disease, nonfatal myocardial infarction, fatal or nonfatal isch- The current place in therapy for PCSK9 inhibitors is in addi-
emic stroke, or unstable angina requiring hospitalization. At tion to maximally tolerated LDL cholesterol lowering therapy
median follow-up of almost 3 years, alirocumab significantly (e.g., maximally tolerated statin and ezetimibe) for patients
reduced the composite primary end point, with the greatest with heterozygous or homozygous FH or for patients with
absolute benefit among patients with a baseline LDL choles- clinical ASCVD who require additional LDL cholesterol lower-
terol greater than 100 mg/dL. ing (Grundy 2019). The use of PCSK9 inhibitors is also recom-
The FOURIER trial evaluated the second PCSK9 inhib- mended for patients with statin intolerance and high ASCVD
itor, evolocumab, in patients with clinical ASCVD plus addi- risk. No RCTs have evaluated the strategy of adding ezetimibe
tional risk factors and who had an LDL cholesterol 70 before a PCSK9 inhibitor.
mg/dL or greater or non-HDL cholesterol 100 mg/dL or greater
on maximally tolerated statin therapy with or without ezeti- Omega-3 Fatty Acids
mibe (Sabatine 2017). The median baseline LDL cholesterol Very low-density lipoprotein cholesterol and triglyceride
was 92 mg/dL with 70% of patients on a high-intensity statin, synthesis are inhibited in the liver by O3FAs, which are pre-
and 30% of patients on a moderate-intensity statin (Sabatine dominately used for triglyceride lowering (manufacturer
2017). The composite primary end point was cardiovascu- package insert). The O3FAs are available by prescription,
lar death, myocardial infarction, stroke, hospitalization for such as omega-3-ethyl esters (Lovaza) or icosapent ethyl

PSAP 2022 Book 1 • Cardiology 166 Non-Statin Therapy for Dyslipidemia


Patient Care Scenario
A 51-year-old African American woman is referred to your principal. Today her blood pressure is 139/85 mm Hg,
pharmacotherapy clinic for review of her ASCVD risk. She heart rate is 78 bpm, and BMI is 33.2 kg/m2. Her most
has mild myalgia associated with statin therapy after recent fasting lipid panel is as follows: total cholesterol
failure of 4 alternative statins and her current statin at a 160 mg/dL, HDL cholesterol 45 mg/dL, LDL cholesterol
higher dose. Her current diagnoses are hypertension, type 85 mg/dL, and triglycerides 149 mg/dL. Based on the
2 diabetes, and obesity, and her medical history is sig- American Association of Clinical Endocrinologists and
nificant for preeclampsia. She has no family history of American College of Endocrinology recommendations,
premature ASCVD. She denies use of tobacco and reports which of the following is the best recommendation for this
drinking 1 standard alcoholic beverage 2–3 times weekly. patient’s lipid-lowering therapy?
She reports full adherence to the following medications: A. No change to current therapy
amlodipine 5 mg/day, candesartan 16 mg/day, rosuvasta- B. Change rosuvastatin to 10 mg/day and continue current
tin 5 mg/day, and metformin 2000 mg/day. therapy
The patient eats a typical Western diet and is gener- C. Add ezetimibe to current therapy
ally inactive physically in her job as a middle school vice
D. Add PCSK9 inhibitor to current therapy
ANSWER
The patient’s 10-year ASCVD risk score is 12.4% and she increasing the rosuvastatin dose is not the best option
has type 2 diabetes. Therefore, she is indicated, at the because her previous management has failed with sev-
very least, for a moderate-intensity statin. According to eral statins, including rosuvastatin at a dose higher than
the 2018 American Heart Association/American College 5 mg/day. Still, a change is warranted. According to the
of Cardiology blood cholesterol guidelines, rosuvastatin 2020 American Association of Clinical Endocrinologists/
5 mg/day is a moderate-intensity statin. According to the American College of Endocrinology guidelines her goal
same guidelines to determine statin therapy for a patient LDL cholesterol is less than 70 mg/dL because of her
with diabetes, further risk assessment is warranted to many risk factors. Adding ezetimibe will reach this goal
assess the potential benefit of additional lipid-lowering and is the best option because data supporting the use of
therapy. The patient has several risk factors for ASCVD, PCSK9 inhibitors for primary prevention without markedly
including hypertension, obesity, and a history of pre- elevated LDL cholesterol are scant; therefore, the greater
eclampsia (a risk enhancing factor). Therefore, the patient expense and injectable route are not justified.
is candidate for high-intensity statin therapy. However,

1. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APHA/ASPC/NLA/PCNA guideline on the
management of blood cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on clinical
practice guidelines. Circulation 2019;139:e1082-143.
2. Handelsman Y, Jellinger PS, Guerin CK, et al. Consensus statement by the American Association of Clinical Endocrinologists and
American College of Endocrinology on the management of dyslipidemia and prevention of cardiovascular disease algorithm—2020
executive summary. Endocr Pract 2020;26:1196-224.

(Vascepa, EPA only) or over-the-counter and contain a combi- patients were using a low-intensity statin (pravastatin 10 mg
nation of EPA and/or DHA. Depending on the dose and base- or simvastatin 5 mg) (Yokoyama 2007). The primary com-
line triglyceride concentration, O3FAs lower triglycerides by posite end point was sudden cardiac death, fatal and nonfa-
10%–50%. The combination products of O3FA with EPA and tal myocardial infarction, and other nonfatal events. Over a
DHA (i.e., omega-3-ethyl esters) may lead to small increases median follow up of 4.6 years, a 19% reduction (ARR 0.7%; HR
in LDL cholesterol, particularly if triglycerides are mark- 0.81; 95% CI, 0.69–0.95) in the primary composite end point
edly elevated (e.g., ≥ 500 mg/dL) (Feingold 2000), whereas was observed with EPA 1.8 g/day plus background statin ther-
O3FA combination products with EPA only (i.e., icosapent
apy compared with statin monotherapy (Yokoyama 2007).
ethyl) have not been shown to increase LDL cholesterol. The
Among those with preexisting coronary artery disease, a 19%
O3FA products sold as a food supplement do not have FDA
reduction in major coronary events was observed, compared
approval, and the amount of EPA and DHA varies; therefore,
with patients only using a statin (ARR 2%; HR 0.81; 95% CI,
these agents are not preferred.
0.66–1.00).
Several trials of low-dose O3FA have failed to demonstrate
an ASCVD benefit. However, several trials, including MARINE, The REDUCE-IT trial, published in 2018, included patients
ANCHOR, JELIS, and REDUCE-IT, used higher doses of EPA age 45 years and older with clinical ASCVD or patients age
and demonstrated an ASCVD benefit when used in combina- 50 years and older with diabetes and other risk factors (Bhatt
tion with statin therapy (Bhatt 2019; Ballantyne 2012; Bays 2019). Other inclusion criteria were triglyceride concentra-
2012;Yokoyama 2007). The JELIS trial was conducted in Jap- tions of 135–499 mg/dL and LDL cholesterol concentration of
anese patients with or without coronary artery disease. The 41–100 mg/dL with stable statin therapy (with or without eze-
median baseline LDL cholesterol was 85 mg/dL and 90% of timibe). The trial found a significant 25% RR reduction (ARR

PSAP 2022 Book 1 • Cardiology 167 Non-Statin Therapy for Dyslipidemia


4.8%, HR 0.75; 95% CI, 0.68–0.83) in the primary composite with triglycerides 500 mg/dL or greater to prevent acute pan-
cardiovascular outcome. creatitis (Grundy 2019). Although effective for triglyceride
Compared with JELIS, the REDUCE-IT trial used a higher lowering, results from clinical trials examining the efficacy of
dose of EPA (icosapent ethyl 4 g/day) in patients already O3FA on cardiovascular risk reduction have been mixed. Sev-
using background statin therapy as well as a placebo group eral clinical trials are ongoing to better understand the asso-
(Bhatt 2019). The primary composite end point of cardiovas- ciation between elevated triglycerides, residual ASCVD risk
cular death, nonfatal myocardial infarction, nonfatal stroke, and O3FA formulations.
coronary revascularization, or unstable angina occurred in
17.2% of patients compared with 22% in the placebo group Ezetimibe
(HR 0.75; 95% CI, 0.68–0.83). Of interest, the results demon- Ezetimibe is a selective inhibitor of the Niemann-Pick C1-Like
strated that the cardiovascular benefit was not specifically 1 protein, which is involved in the absorption of dietary and
related to the triglyceride reduction. However, in light of biliary cholesterol at the brush border of the small intestine,
the overall cardiovascular benefit, icosapent ethyl is rec- per the manufacturer package insert. Inhibition leads to a
ommended by several society guidelines for ASCVD risk decrease in the delivery of dietary cholesterol to the liver.
reduction in patients matching the study population. Both This decreased delivery causes a reduction of hepatic cho-
the JELIS and REDUCE-IT findings are in contrast to those lesterol stores by reducing the production of very low-density
of the STRENGTH trial, which studied an EPA/DHA formu- lipoprotein cholesterol, and subsequently LDL cholesterol.
lation in 13,078 patients with controlled LDL cholesterol, The reduction in hepatic cholesterol also leads to an increase
low HDL cholesterol, and high triglycerides (Nicholls 2020). in the expression of LDL receptors, which promotes LDL
Despite EPA/DHA significantly lowering triglycerides and cholesterol clearance. Ezetimibe lowers LDL cholesterol by
high-sensitivity C-reactive protein, there was no difference 13%–20% and by up to 25% when used in combination with a
in the composite outcome of cardiovascular death, nonfa- statin (Grundy 2019).
tal myocardial infarction, nonfatal stroke, coronary revascu- Evidence for ASCVD risk reduction with ezetimibe has
larization, or unstable angina requiring hospitalization (EPA/ been mixed and difficult to determine. Simvastatin and eze-
DHA group, 12.0% vs. corn oil group 12.2%; HR 0.99; 95% CI, timibe were compared with placebo in two RCTs—the SEAS
0.90–1.09). and SHARP trials (Baigent 2011; Rossebø 2008). In the SEAS
The most common adverse effects of O3FAs are GI (e.g., trial, simvastatin and ezetimibe failed to significantly reduce
abdominal pain, nausea, diarrhea) and a fishy aftertaste, per the composite outcome of combined aortic-valve events and
package inserts. Bleeding risk can be increased in those who ischemic events in patients with aortic stenosis (HR 0.96;
are concomitantly taking anticoagulants or antiplatelets. 95% CI, 0.83–1.12) (Rossebø 2008). However, in the SHARP
Serious bleeding events occurred more often in patients ran- trial, simvastatin plus ezetimibe reduced LDL cholesterol and
domized to icosapent ethyl versus placebo in the REDUCE-IT reduced the primary end point of first major ASCVD events
trial (2.7% vs. 2.1%, p=0.06), as did hospitalizations for atrial compared with placebo over a median follow up of 4.9 years
fibrillation (3.1% vs. 2.1%, p=0.004) (Bhatt 2019). Three addi- in patients with chronic kidney disease (Baigent 2011). Isolat-
tional RCTs suggest a dose-related risk of atrial fibrillation ing the actual effect of ezetimibe in the SHARP trial was dif-
with O3FA supplementation (Albert 2021; Kalstad 2021; Nich- ficult because of the lack of a statin-only comparator group.
olls 2020). At a dose of 4 g/day (EPA/DHA), a statistically sig- In a third trial, ENHANCE, ezetimibe plus simvastatin did
nificant increase in the risk of atrial fibrillation was observed not show a decrease in carotid intima media thickness, a
(Nicholls 2020), although an intermediate dose of 1.8 g/day surrogate marker of ASCVD risk, compared with simvastatin
(EPA/DHA) led to a nonsignificant increase in atrial fibrilla- monotherapy among patients with heterozygous FH (Kaste-
tion risk (Kalstad 2021). Among the available RCTs, the only lein 2008). Finally, in 2015, the IMPROVE-IT trial was the first
one to evaluate incident atrial fibrillation as the primary out- to demonstrate a reduction in ASCVD risk with the addition of
come was the VITAL Rhythm Study, which used O3FA at 840 ezetimibe to simvastatin in patients with recent acute coro-
mg/day (EPA/DHA), and found a nonsignificant increase in nary syndromes (Cannon 2015). Combination therapy demon-
the incidence of atrial fibrillation compared with placebo (HR strated a statistically significant, but clinically modest,
1.09; 95% CI, 0.96–1.24) (Albert 2021). The risk of atrial fibril- reduction in events over 7-year follow-up, without safety con-
lation should be weighed against the benefits of high-dose cerns (HR 0.93; 95% CI, 0.89–0.99) (Cannon 2015). Because
O3FA, and, if used in combination, warrants close monitoring evidence for the use of ezetimibe plus simvastatin in patients
and patient education. with stable clinical ASCVD is lacking, the Food and Drug
Although published before REDUCE-IT, the 2018 American Administration has not approved ASCVD risk reduction as an
Heart Association/American College of Cardiology blood cho- indication for ezetimibe; however, the clinical practice guide-
lesterol clinical practice guideline recommends O3FA (2–4 g/ lines still support the use of ezetimibe use in patients with
day) as an adjunct to lifestyle intervention (e.g., low fat diet, ASCVD, on maximally tolerated statin therapy, and in need of
avoidance of refined carbohydrates, and alcohol) in patients additional LDL cholesterol lowering (Grundy 2019).

PSAP 2022 Book 1 • Cardiology 168 Non-Statin Therapy for Dyslipidemia


Patient Care Scenario
H.G., a 61-year-old African American man with a medi- Part 1
cal history of hypertension, dyslipidemia, prediabetes, Above which one of the following LDL cholesterol thresh-
transient ischemic attack (2 years ago), presents for the olds would it be best to recommend a non-statin therapy
outpatient management of dyslipidemia. His home drugs for H.G.?
include: lisinopril 20 mg/day, indapamide 2.5 mg/day, A. < 130 mg/dL
aspirin 81 mg/day, and atorvastatin 20 mg/day. H.G.’s
B. < 100 mg/dL
most recent lipid panel shows total cholesterol 170 mg/
C. < 70 mg/dL
dL, high-density lipoprotein cholesterol (HDL cholesterol)
41 mg/dL, low-density lipoprotein cholesterol (LDL cho- D. < 55 mg/dL
lesterol) 96 mg/dL, and triglycerides 165 mg/dL. His most
recent blood pressure is 129/79 mm Hg.
ANSWER
Less than 70 mg/dL is the best option for this patient (Answer B), would be reasonable for some patients with-
because he has ASCVD as noted by his previous tran- out ASCVD. Less than 55 mg/dL (Answer D), would be
sient ischemic attack (Answer C is correct). Less than reasonable for premature ASCVD, progressive ASCVD, or
130 mg/dL (Answer A), is not reasonable for someone with ASCVD with multiple or substantial other risk factors.
ASCVD or risk factors for ASCVD. Less than 100 mg/dL

1. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APHA/ASPC/NLA/PCNA guideline on the
management of blood cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on clinical
practice guidelines. Circulation 2019;139:e1082-143.
2. Handelsman Y, Jellinger PS, Guerin CK, et al. Consensus statement by the American Association of Clinical Endocrinologists and
American College of Endocrinology on the management of dyslipidemia and prevention of cardiovascular disease algorithm—2020
executive summary. Endocr Pract 2020;26:1196-224.
Part 2 A. Add ezetimibe.
H.G.’s care team determines that additional LDL choles- B. Add evolocumab.
terol lowering is desired. Which one of the following is C. Increase atorvastatin dose.
best to recommend to lower H.G.’s LDL cholesterol below D. Increase atorvastatin dose and add ezetimibe.
his threshold?
ANSWER
Because the patient is not taking a high-intensity statin, threshold. But there is no such history (Answer B is incor-
although is indicated for one based on the presence of rect). Therefore, optimizing this patient’s statin dose and
ASCVD (his prior transient ischemic attack), the best first adding ezetimibe is the best option (Answer D is cor-
option is to increase his atorvastatin dose; however, this rect). If the patient was taking an optimized statin dose,
alone will likely still leave him above his LDL cholesterol but needed more than 25% additional LDL cholesterol
threshold (Answer C is incorrect). If there was a confirmed reduction to reach goal, then a PCSK9 inhibitor could be
history of statin intolerance, including trials of multiple considered. Answer A and Answer B are incorrect because
other statins including atorvastatin at a higher dose, then they do not increase the atorvastatin dose. Answer C is
a PCSK9 inhibitor could be considered as ezetimibe alone not correct because it is unlikely to achieve the LDL cho-
would not likely lower LDL cholesterol below the desired lesterol threshold.

1. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APHA/ASPC/NLA/PCNA guideline on the
management of blood cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on clinical
practice guidelines. Circulation 2019;139:e1082-143.
2. Handelsman Y, Jellinger PS, Guerin CK, et al. Consensus statement by the American Association of Clinical Endocrinologists and
American College of Endocrinology on the management of dyslipidemia and prevention of cardiovascular disease algorithm—2020
executive summary. Endocr Pract 2020;26:1196-224.

Adverse effects are uncommon with use of ezetimibe. In ezetimibe can be the preferred initial non-statin therapy
clinical trials, the incidence of adverse effects and discontinua- because of its expected benefit on ASCVD outcomes, mod-
tions rates have been similar to placebo (manufacturer’s pack- est LDL cholesterol lowering, tolerability, cost, and once-daily
age insert). Adverse effects occurred in clinical trials in 2% or dosing (Grundy 2019). It is therefore reasonable to consider
less of patients, including fatigue, abdominal pain/diarrhea, adding ezetimibe to maximally tolerated statin therapy in
upper respiratory tract infections, arthralgia, and back pain. patients with clinical ASCVD and an LDL cholesterol of 70
Although outcomes from using combination therapy with mg/dL or greater. Additional factors that place patients with
ezetimibe on top of maximally tolerated statin therapy in clinical ASCVD at very high risk and may favor adding ezeti-
stable clinical ASCVD patients are not extensively studied, mibe to a statin include history of several major ASCVD events

PSAP 2022 Book 1 • Cardiology 169 Non-Statin Therapy for Dyslipidemia


or one major ASCVD event and several high-risk comorbidi- Bempedoic acid has been associated with hyperuricemia
ties, such as diabetes, hypertension, chronic kidney disease, and gout, particularly among patients with a history of gout.
current smoking, persistent LDL cholesterol elevation, and Gout event rates in CLEAR Harmony were 1.2% versus 0.3%
history of heart failure (Grundy 2019). In addition, ezetimibe (p=0.03) and in the Clear Wisdom trial were 2.1% versus 0.8%
therapy is reasonable in patients age 20–75 years with an (p value not reported) (Goldberg 2019; Ray 2019). Bempe-
LDL cholesterol 190 mg/dL or greater who do not achieve at doic acid has also been associated with an increased risk of
least a 50% reduction in LDL cholesterol on maximally toler- tendon rupture, particularly among patients age 60 years or
ated statin therapy and/or have an LDL cholesterol concen- older, those taking a corticosteroid or fluoroquinolone, and in
tration of 100 mg/dL or greater (Grundy 2019). Ezetimibe can those with renal failure or previous tendon disorders.
also be considered as the initial therapy in patients with true Although consistent reductions in LDL cholesterol have
statin intolerance. been observed with bempedoic acid monotherapy or in com-
bination with background lipid-lowering therapy in phase
Bempedoic Acid 3 trials, the effect on cardiovascular morbidity and mor-
Bempedoic acid inhibits adenosine triphosphate-citrate tality has yet to be determined. The CLEAR Outcomes trial
lyase (ACL), which is an enzyme upstream of 3-hydroxy- (NCT02993406, expected to be completed in 2022), is enroll-
3-methyl-glutaryl-coenzyme A reductase in the cholesterol ing patients at high risk for cardiovascular disease and who
biosynthesis pathway (manufacturer’s package insert). have statin intolerance. The primary composite end point
Bempedoic acid is a prodrug that requires coenzyme A acti- includes time from randomization to the first occurrence
vation in the liver. Inhibition of ACL results in decreased cho- of nonfatal myocardial infarction, nonfatal stroke, coronary
lesterol synthesis in the liver, which lowers LDL cholesterol revascularization, or cardiovascular death. Despite the lack of
through up-regulation of LDL receptors. Bempedoic acid also data on ASCVD risk reduction, bempedoic acid was granted
decreases non-HDL cholesterol, apolipoprotein B, and total FDA approval in 2020 as an adjunct to diet and maximally tol-
cholesterol. As a prodrug, bempedoic acid is converted in erated statin therapy for the treatment of adults with heterozy-
the liver, so no active metabolites are present in the skeletal gous FH (e.g., primary prevention) or established ASCVD (e.g.,
muscles, making it a promising alternative for patients with secondary prevention) who require additional lowering of LDL
statin-associated muscle symptoms. Bempedoic acid is also cholesterol. Although the place of bempedoic acid in therapy
available in combination with ezetimibe, which lowers LDL has yet to fully be determined, it provides an additional thera-
cholesterol by 48% compared with 23% with bempedoic acid peutic option for LDL cholesterol lowering in statin-intolerant
monotherapy (Thompson 2016; Ballantyne 2013). patients, those requiring additional LDL cholesterol reduction
In a phase 3 trial (CLEAR Harmony), patients with clini- despite maximally tolerated statin therapy, and those who are
cal ASCVD, heterozygous FH, or both were randomized to unable to afford a PCSK9 inhibitor.
bempedoic acid 180 mg/day or placebo for 52 weeks (Ray
2019). Patients included those stable on maximally tolerated Bile Acid Sequestrants
statins alone or in combination with other non-statin thera- Under normal physiologic conditions, most bile acids are
pies. About 8% of patients were on ezetimibe and 4% were secreted into the small intestine from the liver and reabsorbed
on a fibrate in addition to statin therapy. Results from CLEAR through enterohepatic recirculation. Bile acid sequestrants
Harmony demonstrated that bempedoic acid did not lead to (BAS) (e.g., cholestyramine, colestipol, colesevelam) form a
a higher incidence of adverse events versus placebo, but did non-absorbed resin/bile acid complex in the small intestine
significantly lower LDL cholesterol by an additional 18% ver- that is subsequently excreted in the feces (manufacturers’
sus placebo. Across other phase 3 trials conducted in the package inserts). As bile acids decrease, the hepatic enzyme,
CLEAR program, observed LDL cholesterol reductions with cholesterol 7-α-hydroxylase, is up-regulated, increasing con-
bempedoic acid have been between 14%–40% when added version of cholesterol to bile acid in the liver, which decreases
to background lipid-lowering therapy. The greatest reduc- intracellular cholesterol concentrations. In addition, as cho-
tions in LDL cholesterol were shown when added to ezetimibe lesterol concentrations decrease, LDL receptor uptake is
(Thompson 2016). increased allowing for additional LDL cholesterol clear-
The CLEAR Serenity trial assessed the efficacy and ance. Depending on the dose, BAS reduce LDL cholesterol by
safety of bempedoic acid 180 mg/day compared with pla- 15%–30% when added to statin therapy (Grundy 2019).
cebo in statin-intolerant patients over 24 weeks (Laufs 2019). Three RCTs have evaluated the efficacy of cholestyramine
About 8.4% of patients continued low-dose statin therapy. At for ASCVD risk reduction, although results have been incon-
12 weeks, bempedoic acid demonstrated a 21.4% reduction in clusive (Watts 1992; Brensike 1984; Lipid Research Clin-
LDL cholesterol versus placebo. The most common muscle- ics 1984). Among these three trials, only one demonstrated
related adverse events were myalgia and occurred in 4.7% of a modest reduction in ASCVD risk with BAS monother-
patients using bempedoic acid versus 7.2% of patients in the apy (Lipid Research Clinics 1984). The LRC-CPPT trial ran-
placebo group. domly assigned 3806 men younger than 60 years without

PSAP 2022 Book 1 • Cardiology 170 Non-Statin Therapy for Dyslipidemia


cardiovascular disease but with a total cholesterol of at least therapy. Finally, BAS may be used in pregnant women at high
265 mg/dL to cholestyramine 24 g/day or placebo for a mean ASCVD risk, although monitoring for vitamin K deficiency may
of 7.4 years. The cholestyramine group experienced a 19% be required (Goldberg 2011).
reduction in risk in the primary end point of coronary heart
disease death and/or definite nonfatal myocardial infarction Fibrates
(OR 0.81, 95% CI, 0.70–1.02; p=0.07) (Lipid Research Clinics Fibrates, such as gemfibrozil, fenofibrate, and fenofibric acid,
1984). To date, the effects of colesevelam and colestipol on are derivatives of fibric acid and act as ligands for the nuclear
ASCVD risk reduction have not been determined because hormone transcription factor peroxisome proliferator-activated
no adequately powered trials have explored their impact on receptor α (PPAR-α) (manufacturers’ package inserts). The
reducing ASCVD risk. PPARs regulate lipid metabolism that function clinically to
Given the inconclusive findings across trials exploring decrease triglyceride concentrations by 20%–50% through
impact of BAS on ASCVD risk reduction, several secondary increased expression of lipoprotein lipase and decreasing
analyses have been conducted. In a systematic review and apolipoprotein CII concentration. Fenofibrate is more effec-
meta-analysis of RCTs of cholestyramine and colesevelam, tive than gemfibrozil in lowering triglyceride levels. Fibrates
Mendelian randomized was used to estimate the effect of BAS also increase HDL cholesterol by increasing the expression of
on reducing coronary artery disease risk (Ross 2015). Both apolipoprotein AI and apolipoprotein AII, and have also been
cholestyramine and colesevelam reduced LDL cholesterol and shown to be effective at reducing non-HDL cholesterol.
were associated with a reduction in the risk of coronary artery The Helsinki Heart Study (HHS) compared gemfibrozil 600
disease. In a second systematic review and meta-regression mg twice daily versus placebo for 5 years in middle-age men
analysis, the use of statin and non-statin therapies that act without clinical ASCVD, and found a 34% reduction in the pri-
by up-regulation of LDL cholesterol receptor expression to mary end point of coronary heart disease events (myocar-
reduce LDL cholesterol (e.g., BAS) were associated with simi- dial infarction and cardiovascular death) (Frick 1987). In an
lar RR reductions of major vascular events per change in LDL 18-year follow up of the HHS, those who received gemfibrozil
cholesterol; the RR for major vascular events per 38.7-mg/dL had a 23% lower RR of coronary heart disease mortality com-
reduction in LDL cholesterol was 0.77 for statins and 0.75 for pared with those in the placebo group (Tenkanen 2006). Those
non-statins working primarily through LDL receptor up-reg- in the highest tertile of BMI and triglycerides experienced the
ulation (Silverman 2016). Although the use of BAS is effec- greatest benefit. However, in 2010, results from the ACCORD-
tive at lowering LDL cholesterol, data supporting ASCVD risk Lipid trial demonstrated that fenofibrate and simvastatin did
reduction with BAS remains limited. not reduce ASCVD risk in patients with type 2 diabetes com-
No systemic absorption occurs with BAS; therefore, they pared with simvastatin monotherapy (Ginsberg 2010). Simi-
do not cause systemic adverse effects (Grundy 2019). The lar results were observed in a 5-year follow-up study of the
most common adverse effects are GI, including constipa- ACCORD-Lipid trial (Elam 2017). However, in subgroup anal-
tion, bloating, flatulence (manufacturers’ package inserts). yses of patients in the ACCORD-Lipid trial with triglycerides
Comparatively, colesevelam has the lowest GI adverse in the highest third (≥ 204 mg/dL) and HDL cholesterol in the
effect profile in the class. Of note, BAS may raise triglyceride lowest third (≤ 34 mg/dL), lower cardiovascular event rates
levels and should be avoided if fasting hypertriglyceridemia were observed with the addition of fenofibrate to simvasta-
(> 300 mg/dL) is present (Grundy 2019). Limitations to using tin compared with simvastatin monotherapy (Ginsberg 2010).
BAS include pill burden (most formulations require admin- The most common adverse effects of fibrates are mild
istering several tablets dosed twice daily), and potential for GI disturbances that lessen with time. Because fibrates
drug–drug interactions (e.g., digoxin, warfarin, and thyroid increase biliary cholesterol excretion, the risk of gallstone
hormone). Also, BAS may impair the absorption of fat-soluble formation is increased. Muscle-related toxicity (e.g., myopa-
vitamins (e.g., A, D, E, K), and should be administered at least thy and rhabdomyolysis) has been reported in patients taking
1 hour before or at least 4 hours after other medications to fibrate monotherapy, and this risk is increased when com-
limit potential for drug–drug interactions (manufacturers’ bined with a statin (Wiggins 2016). However, muscle-related
package inserts). Potential for drug–drug interactions are toxicity occurs more often with gemfibrozil versus fenofi-
less with colesevelam compared with cholestyramine and brate in combination with a statin (Amend 2011; Jones 2005).
colestipol (Grundy 2019). The FDA-approved product labeling for simvastatin states
Patients who are age 20–75 years can be treated with a that gemfibrozil is contraindicated and recommends avoid-
BAS when baseline LDL cholesterol is 190 mg/dL or greater ing use of gemfibrozil with all other available statins. How-
and a 50% reduction in LDL cholesterol is not achieved with ever, on the basis of differences in pharmacokinetic profiles
maximally tolerated statin therapy and ezetimibe (Grundy of the statins, a 2016 scientific statement from the Amer-
2019). In addition, BAS can serve as an alternative agent for ican Heart Association recommended that gemfibrozil be
patients who are intolerant to ezetimibe requiring additional avoided in combination with lovastatin, pravastatin and sim-
LDL cholesterol lowering beyond maximally tolerated statin vastatin—and may be acceptable to use in combination with

PSAP 2022 Book 1 • Cardiology 171 Non-Statin Therapy for Dyslipidemia


atorvastatin, rosuvastatin, and pitavastatin if clinically indi- inclisiran (ORION 4, NCT03705234) began enrolling subjects
cated, and if fenofibrate (or fenofibric acid) is not an option in 2018, with results expected in 2025. Inclisiran was denied
(Wiggins 2016). Fibrates should not be used in patients with FDA approval in 2020 because of facility inspection-related
severe hepatic or renal dysfunction and in patients with pre- conditions. However, inclisiran gained approval in the Euro-
existing gallbladder disease or biliary cirrhosis (manufactur- pean Union in 2020 for adults as an adjunct to diet  in com-
ers’ package inserts). bination with maximally tolerated statin therapy in patients
Fibrates are primarily used in the treatment of hypertri- who cannot reach LDL cholesterol goals. Inclisiran is also
glyceridemia when triglycerides are greater than 500 mg/dL approved in the European Union for use alone or in combina-
to prevent acute pancreatitis in combination with a statin tion for patients who are statin-intolerant.
(Grundy 2019). Evidence does not support the use of fibrates
for ASCVD risk reduction, especially when used in combina- Pemafibrate
tion with statin therapy. Fibrates may provide ASCVD risk Through selective activation of the peroxisome proliferator-
benefit in certain patients (e.g., those with type 2 diabetes), activated receptors, pemafibrate is an advancement com-
or in those with certain patterns of dyslipidemia (e.g., high pared with older fibrates, which have less potent activity and
triglycerides and low HDL cholesterol), but evidence remains limited efficacy. Several trials have demonstrated greater
uncertain (Sisson 2018). If used in combination with a statin, triglyceride reductions with pemafibrate compared with
fenofibrate is preferred to gemfibrozil because of the lower fenofibrate, with less frequent adverse events (Arai 2018;
risk of muscle related toxicity (Grundy 2019). Ishibashi 2018). Ongoing clinical trials are evaluating pema-
fibrate to reduce residual cardiovascular risk in patients
Niacin using statins (Pradhan 2018) and in patients with fasting
Niacin primarily lowers triglycerides, raises HDL cholesterol, hypertriglyceridemia and renal impairment (NCT03011450,
and has mild LDL cholesterol lowering (Grundy 2019). On the NCT03001817).
basis of unfavorable results from the AIM-HIGH and HPS2-
THRIVE trials (Landray 2014; Boden 2011), current evidence Volanesorsen
does not support the routine use of niacin in light of its lack of
Volanesorsen is an antisense oligonucleotide inhibitor of
efficacy in reducing ASCVD risk, potential adverse effects and
apolipoprotein C-III mRNA, leading to its degradation and
harms, such as flushing, hyperglycemia, and hepatoxicity.
the reduction in its synthesis. Synthesized primarily in the
liver, apolipoprotein C-III is a component of triglyceride lipo-
EMERGING NON-STATIN THERAPIES: proteins, thus playing a role in regulating plasma triglyceride
MEDICATIONS IN THE PIPELINE concentrations (Ooi 2008). Increased levels of apolipopro-
tein C-III have been associated with impaired lipolysis and
Inclisiran
reductions in the clearance of triglyceride lipoprotein as well
The package insert for inclisiran describes it as a small inter-
as hepatic lipase activity (Ginsberg 1986). In addition, apo-
fering ribonucleic acid that inhibits PCSK9 synthesis in the
lipoprotein C-III has been independently associated with an
liver through directing catalytic breakdown of mRNA for
increased risk for cardiovascular disease (Mendivil 2011;
PCSK9. This action increases LDL cholesterol receptor recy-
Sacks 2000).
cling, which increases LDL cholesterol uptake and lowers LDL
In a meta-analysis of phase 2 and phase 3 clinical studies
cholesterol. On average, inclisiran lowers LDL cholesterol by
of volanesorsen, significant reductions in very low-density
50%–55% with twice yearly dosing. Patients in the ORION 9,
lipoprotein cholesterol, triglycerides, and apolipoprotein
10 and 11 trials required additional LDL cholesterol reduction
C-III, and increases in HDL cholesterol and LDL cholesterol
despite taking a maximally tolerated statin dose with or with-
have been observed (Fogacci 2020). Although a signifi-
out ezetimibe (Raal 2020; Ray 2020). ORION 9 assessed the
cant increase in LDL cholesterol was observed, the baseline
efficacy of inclisiran in patients with heterozygous FH (Raal
LDL cholesterol was very low (about 28 mg/dL) and did not
2020). Inclisiran significantly reduced the mean percentage
increase to more than 70 mg/dL. However, volanesorsen was
change in LDL cholesterol by 48% compared with placebo.
rejected by the FDA in 2018 because of concerns related to
Both ORION-10 and ORION-11 included patients with clini-
thrombocytopenia and subsequent bleeding, although it was
cal ASCVD and ASCVD risk equivalents, and demonstrated a
approved in 2019 in the European Union for familial chylomi-
significant reduction in the mean percentage change in LDL
cronemia syndrome.
cholesterol by 52% and 50%, respectively (Ray 2020). Incli-
siran also significantly reduced the time-adjusted percent-
Evinacumab
age change in LDL cholesterol from baseline by 54% and 49%
Evinacumab is an antagonist of angiopoietin-like protein 3,
in ORION 10 and 11, respectively. The only adverse reactions
which inhibits hydrolysis of triglycerides by lipoprotein lipase
associated with inclisiran in clinical trials were injection site
(Arca 2020; Ruscica 2020). Evinacumab reduces triglycerides
reactions. The cardiovascular efficacy and safety trial of

PSAP 2022 Book 1 • Cardiology 172 Non-Statin Therapy for Dyslipidemia


by up 76% and LDL cholesterol by more than 50% with no dif- 2014). The key component to making a lifestyle intervention
ferences in adverse events compared with placebo (Arca “high-intensity” is the frequency of patient contact, at least
2020; Rosenson 2020; Gaudet 2017; Dewey 2017). According 14 visits over 6 months (individually or in groups, in person or
to the manufacturer package insert, evinacumab-dgnb injec- remote). At least monthly contact thereafter can help main-
tion was approved in 2021 for homozygous familial hypercho- tain changes (Jensen 2014). Some programs include even
lesterolemia treatment in patients age 12 years and older. more frequent patient contact, such as the National Diabetes
Prevention Program, which meets weekly for 6 months, then
IMPLEMENTING NON-STATIN monthly for 6 months (Ely 2017).
THERAPY
Non-Statin Medications
Therapeutic Lifestyle Change In choosing which non-statin medication to implement, the
The primary non-statin therapy for dyslipidemia is therapeu- strongest consideration must be given to the evidence of car-
tic lifestyle change, ostensibly an approach able to benefit diovascular benefit. However, this concern must be weighed
all patients. As with drug therapy, the principal rationale is with cost to the patient, insurance restrictions, and medica-
to decrease cardiovascular morbidity and mortality. As such, tion administration. For ASCVD risk reduction related to LDL
all core tenants of a healthy lifestyle should be assessed cholesterol, published data on the PCSK9 inhibitor class pro-
and addressed in a patient-specific manner, including the vide the most robust evidence for this indication. Clinicians
following: 1) smoking cessation; 2) dietary modification to
reduce saturated fat intake; decrease intake of highly pro-
cessed, high sodium, and high sugar foods and beverages;
increase fruit and vegetable intake; and moderate calories to Practice Points
lose weight or maintain a healthy weight; 3) decrease alco- • Optimize statin therapy before considering non-statin
hol intake; 4) increase physical activity; and 5) adopt healthy medications.
• The nocebo effect contributes to a substantial proportion
sleep habits (Jacobson 2015b; Kuehn 2019). Specific life-
of statin-related myalgia.
style approaches can be used to lower triglycerides, either • Many patients who cannot tolerate one statin can tolerate
with drug therapy when triglyceride concentration is at least another statin instead.
500  mg/dL or independently for triglyceride concentrations • In patients unable to tolerate the statin intensity for which
of 150–499 mg/dL. When used together, the following life- they are indicated, the maximum tolerated statin should be
style approaches can lower triglycerides by 20%–50%: 1) lim- used.
• Risk reduction for ASCVD is the primary rationale for using
iting drinks high in sugar (especially fructose); 2)  limiting
non-statin medications.
foods high in sugar, particularly those without fiber; 3) • The PCSK9 inhibitors have the strongest data support-
avoiding or limiting alcohol consumption; 4) increasing ing cardiovascular risk reduction, among non-statin
dietary fiber; 5) avoiding trans fats; 6) eating marine-derived medications.
omega-3 polyunsaturated fatty acids; and 7) performing at • In patients already optimized on statin therapy who are
near their LDL cholesterol goal, ezetimibe can be consid-
least moderate-intensity aerobic activity at least 5 days per
ered over PCSK9 inhibitors because of the lower cost and
week (Jacobson 2015b; Miller 2011).
oral administration of ezetimibe.
In implementing therapeutic lifestyle changes, the impor- • Icosapent ethyl should be used to reduce the risk for
tance of a patient-specific approach cannot be overstated. ASCVD in patients with the following criteria: 1) controlled
Cultural and socioeconomic factors play a significant role LDL cholesterol, 2) triglycerides 135–499 mg/dL, and
in what types of food patients may eat and the preferences 3) ASCVD or several cardiovascular risk factors.
• The finding of elevated triglycerides is only a specific
for what types of physical activity the patients will engage
indication for non-statin therapy when the concentration is
in based on physical limitations and work-life/neighbor- 500 mg/dL or greater.
hood factors (American Diabetes Association 2021a; Spen- • Serum triglycerides less than 1000 mg/dL can be treated
cer Bonilla 2016; Jensen 2014). A whole-patient approach with high-intensity statin therapy as a first-line option,
will also benefit patients because comorbidities (e.g., depres- with or without triglyceride-specific agents, to lower tri-
sion, clinical hypothyroidism) and personal factors (e.g., poor glycerides and reduce cardiovascular risk.
• Serum triglyceride concentration of 1000 mg/dL or greater
work-life balance) can impede effective therapeutic lifestyle
should be treated with prescription omega-3-acid ethyl
change. Moreover, social factors, including family behav- esters, icosapent ethyl, or fibrates.
iors, can either enhance or diminish patients’ ability to imple- • Therapeutic lifestyle changes to increase physical
ment therapeutic lifestyle changes (Wang 2014). Although activity and adopt a healthier eating pattern can benefit
these factors can be emphasized over time to help patients all patients.
• To optimize patient success with therapeutic lifestyle
shift toward healthier habits, the strongest evidence sup-
changes, clinicians should provide frequent (up to weekly
ports—in willing patients—implementing these changes as a for 6 months) and robust support to patients.
high-intensity, comprehensive lifestyle intervention (Jensen

PSAP 2022 Book 1 • Cardiology 173 Non-Statin Therapy for Dyslipidemia


and their staff must be prepared to address prior authoriza- American Diabetes Association. 10. Cardiovascular disease
tion paperwork in a timely manner to avoid therapy abandon- and risk management: standards of medical care in diabe-
ment. In addition, patients must be properly counseled and tes—2021. Diabetes Care 2021b;44:S125-50.
willing to administer a subcutaneous injection. Despite the Arai H, Yamashita S, Yokote K, et al. Efficacy and safety of
limitations to PCSK9 inhibitor access and use, their uptake pemafibrate versus fenofibrate in patients with high tri-
in practice has been markedly increasing (Sumarsono 2021). glyceride and low HDL cholesterol levels: a multicenter,
placebo-controlled, double-blind, randomized trial. J Ath-
Ezetimibe, although with lesser evidence of cardiovascu-
eroscler Thromb 2018;25:521-38.
lar benefit, has the advantage of generic availability and oral
administration. Thus, ezetimibe has a substantial access and Arca M, D’Erasmo L, Minicocci I. Familial combined hypolip-
ease of use advantage over PCSK9 inhibitors. Although pro- idemia: angiopoietin-like protein-3 deficiency. Curr Opin
Lipidol 2020;31:41-8.
viding LDL cholesterol reduction similar to that with ezeti-
mibe, the effect of bempedoic acid on ASCVD risk is unknown. Baigent C, Landray MJ, Reith C, et al. The effects of lower-
These points are accentuated given the advantages of eze- ing LDL cholesterol with simvastatin plus ezetimibe in
patients with chronic kidney disease (Study of Heart and
timibe, in addition to ezetimibe’s exceptional tolerability.
Renal Protection): a randomised placebo-controlled trial.
Although less expensive and administered orally, the use of Lancet 2011;377:2181-92.
BAS is limited by adverse effects and chelating drug interac-
tions. Icosapent ethyl essentially stands alone with respect to Ballantyne CM, Bays HE, Kastelein JJ, et al. Efficacy and
safety of eicosapentaenoic acid ethyl ester (AMR101)
medications for ASCVD risk reduction that is not associated
therapy in statin-treated patients with persistent high
LDL cholesterol lowering. As such, guideline-directed use in triglycerides (from the ANCHOR study). Am J Cardiol
the proper patient populations should guide implementation. 2012;110:984-92.
Regarding treatments specifically for very high triglycerides,
Ballantyne CM, Davidson MH, Macdougall DE, et al. Effi-
several options are reasonable and have generic availability, cacy and safety of a novel dual modulator of adenosine
such as omega-3-acid ethyl esters, and fibrates. triphosphate-citrate lyase and adenosine monophos-
phate-activated protein kinase in patients with hyper-
CONCLUSION cholesterolemia: results of a multicenter, randomized,
double-blind, placebo-controlled, parallel-group trial. J Am
The use of non-statin therapy is generally reserved for Coll Cardiol 2013;62:1154-62.
patients already optimized (or thoroughly trialed) on statin
Bays HE, Braeckman RA, Ballantyne CM, et al. Icosapent
therapy. The primary goal of non-statin therapy is ASCVD
ethyl, a pure EPA omega-3 fatty acid: effects on lipopro-
risk reduction, not normalization of the lipid profile or another tein particle concentration and size in patients with very
non-evidence–based surrogate outcome. As such, strong high triglyceride levels (the MARINE study). J Clin Lipidol
preference should be given to selecting agents with RCT 2012;6:565-72.
evidence supporting cardiovascular benefit and using them
Bhatt DL, Steg PG, Miller M, et al. Cardiovascular risk reduc-
clinically aligned with the populations in which they were tion with icosapent ethyl for hypertriglyceridemia. N Engl
studied. Medications to lower triglycerides specifically are J Med 2019;380:11-22.
limited to patients with very high triglyceride concentrations.
Boden WE, Probstfield JL, Anderson T, et al. Niacin in
Therapeutic lifestyle changes have the potential to benefit patients with low HDL cholesterol levels receiving inten-
all patients; however, clinicians must provide robust and fre- sive statin therapy. N Engl J Med 2011;365:2255-67.
quent support to optimize the chances of successfully imple-
Brensike JF, Levy RI, Kelsey SF, et al. Effects of therapy with
menting and maintaining healthy behaviors.
cholestyramine on progression of coronary arterioscle-
rosis: results of the NHLBI type II coronary intervention
REFERENCES study. Circulation 1984;69:313-24.
Albert CM, Cook NR, Pester J, et al. Effect of marine omega-3 Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added
fatty acid and vitamin D supplementation on incident to statin therapy after acute coronary syndromes. N Engl J
atrial fibrillation: a randomized clinical trial. JAMA Med 2015;372:2387-97.
2021;325:1061-73.
Dewey FE, Gusarova V, Dunbar RL, et al. Genetic and pharma-
Amend KL, Landon J, Thyagarajan V, et al. Incidence of hos- cologic inactivation of ANGPTL3 and cardiovascular dis-
pitalized rhabdomyolysis with statin and fibrate use in an ease. N Engl J Med 2017;377:211-21.
insured us population. Ann Pharmacother 2011;45:1230-9.
Elam MB, Ginsberg HN, Lovato LC, et al. Association of feno-
American Diabetes Association. 5. Facilitating behavior fibrate therapy with long-term cardiovascular risk in sta-
change and well-being to improve health outcomes: stan- tin-treated patients with type 2 diabetes. JAMA Cardiol
dards of medical care in diabetes—2021. Diabetes Care 2017;2:370-80.
2021a;44:S53-72.
Ely EK, Gruss SM, Luman ET, et al. A national effort to pre-
vent type 2 diabetes: participant-level evaluation of CDC’s

PSAP 2022 Book 1 • Cardiology 174 Non-Statin Therapy for Dyslipidemia


National Diabetes Prevention Program. Diabetes Care the management of dyslipidemia and prevention of car-
2017;40:1331-41. diovascular disease algorithm—2020 executive summary.
Endocr Pract 2020;26:1196-224.
Feingold KR. Triglyceride lowering drugs. In: Feingold KR,
Anawalt B, Boyce A et al., eds. Endotext. South Dartmouth, Harris WS, Ginsberg HN, Arunakul N, et al. Safety and effi-
MA: MDText.com, 2000–2021. cacy of Omacor in severe hypertriglyceridemia. J Cardio-
vasc Risk 1997;4:385-91.
Fogacci F, Norata GD, Toth PP, et al. Efficacy and safety of
volanesorsen (ISIS 304801): the evidence from phase 2 Herrett E, Williamson E, Brack K, et al. Statin treatment and
and 3 clinical trials. Curr Atheroscler Rep 2020;22:18. muscle symptoms: series of randomised, placebo con-
trolled n-of-1 trials. BMJ 2021;372:n135.
Fonarow GC, van Hout B, Villa G, et al. Updated cost-effec-
tiveness analysis of evolocumab in patients with very Ishibashi S, Arai H, Yokote K, et al. Efficacy and safety
high-risk atherosclerotic cardiovascular disease. JAMA of pemafibrate (K77), a selective peroxisome prolifer-
Cardiol 2019;4:691-5. ator-activated receptor α modulator, in patients with
dyslipidemia: results from a 24-week, randomized, dou-
Frick MH, Elo O, Haapa K, et al. Helsinki Heart Study: prima- ble blind, active-controlled, phase 3 trial. J Clin Lipidol
ry-prevention trial with gemfibrozil in middle-aged men 2018;12:173-84.
with dyslipidemia. Safety of treatment, changes in risk
factors, and incidence of coronary heart disease. N Engl J Jacobson TA, Ito MK, Maki KC, et al. National Lipid Asso-
Med 1987;317:1237-45. ciation recommendations for patient-centered manage-
ment of dyslipidemia: part 1—full report. J Clin Lipidol
Gaudet D, Gipe DA, Pordy R, et al. ANGPTL3 inhibition in 2015a;9:129-69.
homozygous familial hypercholesterolemia. N Engl J Med
2017;377:296-7. Jacobson TA, Maki KC, Orringer CE, et al. National Lipid
Association recommendations for patient-centered man-
Ginsberg HN, Elam MB, Lovato LC, et al. Effects of combina- agement of dyslipidemia: part 2. J Clin Lipidol 2015b;9:S1-
tion lipid therapy in type 2 diabetes mellitus. N Engl J Med 122.e1.
2010;362:1563-74.
Jensen MD, Ryan DH, Apovian CM, et al. 2013 AHA/ACC/
Ginsberg HN, Le NA, Goldberg IJ, et al. Apolipoprotein B TOS guideline for the management of overweight and
metabolism in subjects with deficiency of apolipopro- obesity in adults: a report of the American College of
teins CIII and AI. Evidence that apolipoprotein CIII inhibits Cardiology/American Heart Association task force on
catabolism of triglyceride-rich lipoproteins by lipoprotein practice guidelines and the obesity society. Circulation
lipase in vivo. J Clin Invest 1986;78:1287-95. 2014;129:S102-38.
Giugliano RP, Mach F, Zavitz K, et al. Cognitive func- Jones PH , Davidson MH. Reporting rate of rhabdomyolysis
tion in a randomized trial of evolocumab. N Engl J Med with fenofibrate + statin versus gemfibrozil + any statin.
2017a;377:633-43. Am J Cardiol 2005;95:120-2.
Giugliano RP, Pedersen TR, Park JG, et al. Clinical efficacy Kalstad AA, Myhre PL, Laake K, et al. Effects of n-3 fatty
and safety of achieving very low LDL-cholesterol con- acid supplements in elderly patients after myocardial
centrations with the PCSK9 inhibitor evolocumab: a pre- infarction: a randomized, controlled trial. Circulation
specified secondary analysis of the Fourier trial. Lancet 2021;143:528-39.
2017b;390:1962-71.
Karagiannis AD, Mehta A, Dhindsa DS, et al. How low is safe?
Goldberg AC, Hopkins PN, Toth PP, et al. Familial hypercho- The frontier of very low (<30 mg/dl) LDL cholesterol. Eur
lesterolemia: screening, diagnosis and management of Heart J 2021;42:2154-69.
pediatric and adult patients: clinical guidance from the
National Lipid Association Expert Panel on Familial Hyper- Kastelein JJ, Akdim F, Stroes ES, et al. Simvastatin with or
cholesterolemia. J Clin Lipidol 2011;5:S1-8. without ezetimibe in familial hypercholesterolemia. N Engl
J Med 2008;358:1431-43.
Goldberg AC, Leiter LA, Stroes ESG, et al. Effect of bempe-
doic acid vs placebo added to maximally tolerated statins Kuehn BM. Sleep duration linked to cardiovascular disease.
on low-density lipoprotein cholesterol in patients at high Circulation 2019;139:2483-4.
risk for cardiovascular disease: the clear wisdom random-
ized clinical trial. JAMA 2019;322:1780-8. Landray MJ, Haynes R, Hopewell JC, et al. Effects of extend-
ed-release niacin with laropiprant in high-risk patients. N
Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/ Engl J Med 2014;371:203-12.
AACVPR/AAPA/ABC/ACPM/ADA/AGS/APHA/ASPC/NLA/
PCNA guideline on the management of blood cholesterol: Laufs U, Banach M, Mancini GBJ, et al. Efficacy and safety of
a report of the American College of Cardiology/American bempedoic acid in patients with hypercholesterolemia and
Heart Association Task Force on clinical practice guide- statin intolerance. J Am Heart Assoc 2019;8:e011662.
lines. Circulation 2019;139:e1082-143. The Lipid Research Clinics coronary primary prevention trial
Handelsman Y, Jellinger PS, Guerin CK, et al. Consensus results. I. Reduction in incidence of coronary heart dis-
statement by the American Association of Clinical Endo- ease. JAMA 1984;251:351-64.
crinologists and American College of Endocrinology on

PSAP 2022 Book 1 • Cardiology 175 Non-Statin Therapy for Dyslipidemia


Mendivil CO, Rimm EB, Furtado J, et al. Low-density lipopro- cholesterol and recurrent events (CARE) trial. Circulation
teins containing apolipoprotein c-iii and the risk of coro- 2000;102:1886-92.
nary heart disease. Circulation 2011;124:2065-72.
Schwartz GG, Steg PG, Szarek M, et al. Alirocumab and car-
Miller M, Stone NJ, Ballantyne C, et al. Triglycerides and car- diovascular outcomes after acute coronary syndrome.
diovascular disease: a scientific statement from the Amer- N Engl J Med 2018;379:2097-107.
ican Heart Association. Circulation 2011;123:2292-333.
Silverman MG, Ference BA, Im K, et al. Association between
Newman CB, Preiss D, Tobert JA, et al. Statin safety and lowering LCL-C and cardiovascular risk reduction among
associated adverse events: a scientific statement from the different therapeutic interventions: a systematic review
American Heart Association. Arterioscler Thromb Vasc and meta-analysis. JAMA 2016;316:1289-97.
Biol 2019;39:e38-81.
Sisson EM, Pamulapati L, Bucheit JD, et al. Evolving role
Nicholls SJ, Lincoff AM, Garcia M, et al. Effect of high- of non-statin therapy for the management of dyslipid-
dose omega-3 fatty acids vs corn oil on major adverse emia and cardiovascular risk reduction: past, present, and
cardiovascular events in patients at high cardiovascu- future. Pharmacotherapy 2018;38:164-71.
lar risk: the Strength Randomized Clinical Trial. JAMA
2020;324:2268-80. Spencer Bonilla G, Rodriguez-Gutierrez R , V MM. What we
don’t talk about when we talk about preventing type 2 dia-
Nissen SE, Stroes E, Dent-Acosta RE, et al. Efficacy and tol- betes—addressing socioeconomic disadvantage. JAMA
erability of evolocumab vs ezetimibe in patients with mus- Intern Med 2016;176:1053-4.
cle-related statin intolerance: the GAUSS-3 randomized
clinical trial. JAMA 2016;315:1580-90. Sumarsono A, Lalani HS, Vaduganathan M, et al. Trends in
utilization and cost of low-density lipoprotein cholester-
Ooi EM, Barrett PH, Chan DC, et al. Apolipoprotein C-III: ol-lowering therapies among Medicare beneficiaries: an
understanding an emerging cardiovascular risk factor. Clin analysis from the Medicare part D database. JAMA Cardiol
Sci (Lond) 2008;114:611-24. 2021;6:92-6.

Pradhan AD, Paynter NP, Everett BM, et al. Rationale and Tenkanen L, Mänttäri M, Kovanen PT, et al. Gemfibrozil in
design of the pemafibrate to reduce cardiovascular out- the treatment of dyslipidemia: an 18-year mortality fol-
comes by reducing triglycerides in patients with diabetes low-up of the Helsinki Heart Study. Arch Intern Med
(PROMINENT) study. Am Heart J 2018;206:80-93. 2006;166:743-8.

Raal FJ, Kallend D, Ray KK, et al. Inclisiran for the treatment Thompson PD, MacDougall DE, Newton RS, et al. Treat-
of heterozygous familial hypercholesterolemia. N Engl J ment with etc-1002 alone and in combination with eze-
Med 2020;382:1520-30. timibe lowers LDL cholesterol in hypercholesterolemic
patients with or without statin intolerance. J Clin Lipidol
Ray KK, Bays HE, Catapano AL, et al. Safety and efficacy of 2016;10:556-67.
bempedoic acid to reduce LDL cholesterol. N Engl J Med
2019;380:1022-32. Virani SS, Alonso A, Aparicio HJ, et al. Heart disease and
stroke statistics—2021 update: a report from the American
Ray KK, Wright RS, Kallend D, et al. Two phase 3 trials of Heart Association Circulation 2021;143:e254-743.
inclisiran in patients with elevated LDL cholesterol. N Engl
J Med 2020;382:1507-19. Wang ML, Pbert L, Lemon SC. Influence of family, friend and
coworker social support and social undermining on weight
Rosenson RS, Burgess LJ, Ebenbichler CF, et al. Evinacumab gain prevention among adults. Obesity (Silver Spring)
in patients with refractory hypercholesterolemia. N Engl J 2014;22:1973-80.
Med 2020;383:2307-19.
Watts GF, Lewis B, Brunt JN, et al. Effects on coronary artery
Ross S, D’Mello M, Anand SS, et al. Effect of bile acid disease of lipid-lowering diet, or diet plus cholestyramine,
sequestrants on the risk of cardiovascular events: a men- in the St Thomas’ Atherosclerosis Regression Study
delian randomization analysis. Circ Cardiovasc Genet (STARS). Lancet 1992;339:563-9.
2015;8:618-27.
Wiggins BS, Saseen JJ, Page RL, 2nd, et al. Recommen-
Rossebø AB, Pedersen TR, Boman K, et al. Intensive lipid dations for management of clinically significant drug–
lowering with simvastatin and ezetimibe in aortic stenosis. drug interactions with statins and select agents used in
N Engl J Med 2008;359:1343-56. patients with cardiovascular disease: a scientific state-
Ruscica M, Zimetti F, Adorni MP, et al. Pharmacological ment from the American Heart Association. Circulation
aspects of ANGPTL3 AND ANGPTL4 inhibitors: new thera- 2016;134:e468-e95.
peutic approaches for the treatment of atherogenic dyslip- Wood FA, Howard JP, Finegold JA, et al. N-of-1 trial of a sta-
idemia. Pharmacol Res 2020;153:104653. tin, placebo, or no treatment to assess side effects. N Engl
Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and J Med 2020;383:2182-4.
clinical outcomes in patients with cardiovascular disease. Yokoyama M, Origasa H, Matsuzaki M, et al. Effects of eicos-
N Engl J Med 2017;376:1713-22. apentaenoic acid on major coronary events in hypercho-
Sacks FM, Alaupovic P, Moye LA, et al. VLDL, apolipoproteins lesterolaemic patients (JELIS): a randomised open-label,
B, CIII, and E, and risk of recurrent coronary events in the blinded endpoint analysis. Lancet 2007;369:1090-8.

PSAP 2022 Book 1 • Cardiology 176 Non-Statin Therapy for Dyslipidemia


Self-Assessment Questions
16. A 61-year-old African American man with a medical his- 19. Three weeks later, M.G. returns to clinic complaining of
tory of hypertension, dyslipidemia, prediabetes, and the same pain. She wants to know what else she can do
transient ischemic attack (2 years ago) presents for to lower her cardiovascular risk. Which one of the follow-
the outpatient management of dyslipidemia. His home ing is best to recommend for lowering M.G.’s calculated
drugs include: lisinopril 20 mg/day, indapamide 2.5 ASCVD risk score?
mg/day, aspirin 81 mg/day, and atorvastatin 20 mg/day.
A. Moderate-intensity exercise 150 minutes per week
The patient’s most recent lipid panel shows total choles-
B. Good adherence to a Mediterranean diet
terol 170 mg/dL, HDL cholesterol 41 mg/dL, LDL choles-
C. Successfully quitting smoking
terol 96 mg/dL, and TG 165 mg/dL. His most recent blood
D. Lower blood pressure to 129/78 mm Hg
pressure is 129/79 mm Hg. Which one of the following is
best to recommend for treating this patient’s elevated tri-
Question 20–22 pertain to the following case.
glyceride concentration?
T.C., a 63-year-old African American woman (weight 88.5 kg
A. Icosapent ethyl [195 lb], height 64 inches), is a new patient referred to your
B. Prescription omega-3-acid ethyl esters (O3FAs) lipid assessment and management program. She has trialed
C. Dietary supplement O3FAs and did not tolerate simvastatin, atorvastatin, pravastatin, and
D. Lifestyle change lovastatin because of myalgia. T.C.’s dyslipidemia is currently
being treated with rosuvastatin 5 mg/day after the dose was
Questions 17–19 pertain to the following case reduced from 10 mg/day following leg aches. Her medical his-
M.G. is a 54-year-old non-Hispanic white woman who pres- tory includes type 2 diabetes (A1C 7.2%), myocardial infarction
ents to clinic to follow-up on her lipid panel. She was referred (6 months prior, status post 3 drug-eluting stent placements),
to you for determination of initial therapy and associated hypertension (blood pressure today is 135/82 mm Hg),
counseling. M.G.’s medical history includes: hypertension, chronic kidney disease stage 3 (urinary albumin/creatinine
blood pressure today 130/78 mm Hg; and current cigarette ratio 45 mg/g), former smoker (quit 3 months ago following
smoking 1 pack per day, 35 pack-year history. Results of a myocardial infarction). T.C.’s home drugs include: clopidogrel
lipid panel include: TC 220 mg/dL, HDL cholesterol 31 mg/dL, 75 mg/day, aspirin 81 mg/day, rosuvastatin 5 mg/day, lisino-
TG 265 mg/dL, and LDL cholesterol 136 mg/dL. M.G.’s home pril 40 mg/day, indapamide 2.5 mg/day, metformin 1000 mg
drugs include: losartan 50 mg twice daily, amlodipine 10 twice daily, liraglutide 1.8 mg/day, and empagliflozin 25 mg/
mg/day, and chlortalidone 25 mg/day. Her 10-year athero- day. Her lipid panel after 3 months of rosuvastatin 5 mg/day
sclerotic cardiovascular disease (ASCVD) risk score is 12%. is: TC 231 mg/dL, HDL cholesterol 43 mg/dL, TG 260 mg/dL,
and LDL cholesterol 136 mg/dL.
17. M.G. has agreed to statin therapy. Which one of the fol-
lowing is best to recommend to avoid unnecessary statin 20. Which one of the following is the most likely cause of
discontinuation in M.G.? T.C.’s statin-induced muscle pain?
A. Obtain baseline serum creatinine kinase. A. Vitamin D deficiency
B. Obtain detailed history of muscle pain. B. Co-enzyme Q10 deficiency
C. Start coenzyme Q10 supplementation. C. Genetic predisposition
D. Start vitamin D supplementation. D. Nocebo effect
18. Three weeks after being initiated on atorvastatin 20 mg/ 21. Which one of the following LDL cholesterol thresholds is
day, M.G. returns to clinic. She reports bilateral calf pain recommended by at least one major guideline and offers
and cramping that started 3 days prior; this abated after T.C. the greatest possible ASCVD risk reduction?
her self-discontinuation of atorvastatin. Which one of
A. < 130 mg/dL
the following is best to recommend for M.G.?
B. < 100 mg/dL
A. Retrial atorvastatin 20 mg/day. C. < 70 mg/dL
B. Change to rosuvastatin 10 mg/day. D. < 55 mg/dL
C. Change to ezetimibe 10 mg/day.
D. Change to evolocumab 420 mg once monthly.

PSAP 2022 Book 1 • Cardiology 177 Non-Statin Therapy for Dyslipidemia


22. Which one of the following is best to recommend for Questions 26 and 27 pertain to the following case.

additional LDL cholesterol lowering for T.C.? C.B., a 52-year-old white man (weight 109 kg [240 lb], height
70 inches, waist 41 inches), presents to your clinic for man-
A. Increase rosuvastatin dose.
agement of dyslipidemia. His medical history includes osteo-
B. Add evolocumab.
arthritis of the right knee, generalized anxiety disorder, and
C. Add ezetimibe.
hypertension (blood pressure 132/78 mm Hg). C.B.’s home
D. Add bempedoic acid.
drugs include acetaminophen 650 mg four times daily, escit-
alopram 20 mg/day, amlodipine 10 mg/day, and lisinopril
Questions 23 and 24 pertain to the following case
20 mg/day. He reports having trouble paying for brand name
A primary care physician contacts you to reassess lipid medications because of the more expensive co-pay. C.B.’s
therapy in F.B., a 57-year-old white man (weight 110.5 kg most recent lipid panel results include: TC 220 mg/dL, HDL
[243 lb], height 69 inches) with a history of hypertension cholesterol 42 mg/dL, LDL cholesterol 145 mg/dL, and TG
(125/72 mm Hg taking losartan 50 mg twice daily), type 2 dia- 165 mg/dL. His ASCVD 10-year risk is 7%.
betes (A1C 6.8%, taking metformin 1000 mg twice daily), and
26. According to the American Association of Clinical Endo-
dyslipidemia. F.B.’s baseline lipid panel reveals TC 239 mg/dL,
crinologist and American College of Endocrinology,
HDL cholesterol 32 mg/dL, LDL cholesterol 165 mg, and TG
which one of the following LDL cholesterol goals is best
210 mg/dL. His baseline 10-year ASCVD risk was 23.3% After
to recommend for C.B.?
3 months of therapy with atorvastatin 80 mg, F.B.’s LDL cho-
lesterol is 84 mg/dL. Patient report, pill count, and refill his- A. < 130 mg/dL
tory indicate adherence to atorvastatin therapy. B. < 100 mg/dL
C. < 70 mg/dL
23. Which one of the following LDL cholesterol thresholds is
D. < 55 mg/dL
recommended by at least one major guideline and offers
F.B. the greatest possible ASCVD risk reduction? 27. C.B. has heard so many negative things about statins
from his friends that he refuses to even trial one. Which
A. < 130 mg/dL
one of the following is best to recommend to help C.B.
B. < 100 mg/dL
lower his LDL cholesterol?
C. < 70 mg/dL
D. < 55 mg/dL A. Lifestyle intervention
B. Evolocumab and lifestyle intervention
24. F.B.’s physician requests your opinion on adding a med-
C. Ezetimibe and lifestyle intervention
ication for additional LDL cholesterol lowering. Which
D. Gemfibrozil and lifestyle intervention
one of the following is best to recommend for F.B.?
A. Add ezetimibe.
Questions 28 and 29 pertain to the following case.
B. Add alirocumab.
J.B. is a 59-year-old woman being discharged from the hospi-
C. Add a bile acid sequestrant.
tal after a myocardial infarction (status post two drug-eluting
D. Maintain current regimen.
stent placements). Her medical history includes a previous
25. A 47-year-old African American man (weight 106.8 kg myocardial infarction 2 years ago (status post drug-eluting
[235 lb], height 72 inches) with no contributory med- stent placement), type 2 diabetes (A1C 8.3%), hypertension
ical history is establishing care in your clinic. He is a (blood pressure 134/82 mm Hg), rheumatoid arthritis, hypo-
never smoker and his baseline fasting laboratory values thyroidism, and depression. J.B.’s home drugs include: aspi-
and vital signs are: blood pressure 118/75 mm Hg, TC rin 81 mg/day, ticagrelor 90 mg twice daily, candesartan 32
190 mg/dL, HDL cholesterol 39 mg/dL, reflex direct LDL mg/day, amlodipine 10 mg/day, indapamide 2.5 mg/day, top-
cholesterol 142 mg/dL, TG 710 mg/dL, and blood sugar ical diclofenac gel four times/day, methotrexate 20 mg once
102 mg/dL. The physician asks you for a recommenda- weekly, levothyroxine 100 mcg/day, escitalopram 10 mg/day,
tion to manage the patient’s dyslipidemia. Which one of liraglutide 1.8 mg/day, empagliflozin 25 mg/day, atorvastatin
the following is best to recommend for this patient? 80 mg/day, ezetimibe 10 mg/day. Results of her current lipid
A. Fenofibrate 130 mg/day panel include: TC 115 mg/dL, HDL cholesterol 39 mg/dL, LDL
B. Prescription O3FA 4 g/day cholesterol 68 mg/dL, and TG 41 mg/dL.
C. Icosapent ethyl 2g twice daily
D. Atorvastatin 80 mg/day

PSAP 2022 Book 1 • Cardiology 178 Non-Statin Therapy for Dyslipidemia


28. Which one of the following LDL cholesterol goals is best 30. A 59-year-old man has a medical history that includes
to recommend for J.B. to optimize her risk for another dyslipidemia, peripheral artery disease, and hyperten-
myocardial infarction? sion. The patient is statin intolerant and cannot afford
proprotein convertase subtilisin/kexin type 9 (PCSK9)
A. < 130 mg/dL
inhibitor therapy. His home drugs include lisinopril 20
B. < 100 mg/dL
mg/day, aspirin 81 mg/day, and ezetimibe 10 mg/day.
C. < 70 mg/dL
The patient’s current lipid profile is: TC cholesterol 210
D. < 55 mg/dL
mg/dL, HDL cholesterol 42 mg/dL, reflex direct LDL cho-
29. Which one of the following is best to recommend to opti- lesterol 155 mg/dL, and TG 550 mg/dL. A clinical trial of
mize J.B.’s lipids? which one of the following medications is most likely to
A. Bempedoic acid benefit this patient?
B. Evolocumab A. Evinacumab
C. Cholestyramine B. Volanesorsen
D. No additional pharmacotherapy C. Pemafibrate
D. Inclisiran

PSAP 2022 Book 1 • Cardiology 179 Non-Statin Therapy for Dyslipidemia


Learner Chapter Evaluation: Non-Statin Therapy for Dyslipidemia

As you take the posttest for this chapter, also evaluate the 25. The learning assessment activities used in the chapter
material’s quality and usefulness, as well as the achievement were effective.
of learning objectives. Rate each item using this 5-point scale: 26. The chapter was effective overall.

• Strongly agree 27. The activity met the stated learning objectives.
• Agree 28. If any objectives were not met, please list them here.
• Neutral
• Disagree OTHER COMMENTS
• Strongly disagree
29. Please provide any specific comments related to any
16. The content of the chapter met my educational needs. perceptions of bias, promotion, or advertisement of
commercial products.
17. The content of the chapter satisfied my expectations.
30. Please expand on any of your above responses, and/or
18. The author presented the chapter content effectively.
provide any additional comments regarding this chapter:
19. The content of the chapter was relevant to my practice
and presented at the appropriate depth and scope.
Questions 31–33 apply to the entire learning module.
20. The content of the chapter was objective and balanced.
31. How long did it take you to read the instructional materi-
21. The content of the chapter is free of bias, promotion, and
als in this module?
advertisement of commercial products.
32. How long did it take you to read and answer the assess-
22. The content of the chapter was useful to me.
ment questions in this module?
23. The teaching and learning methods used in the chapter
33. Please provide any additional comments you may have
were effective.
regarding this module:
24. The active learning methods used in the chapter were
effective.

PSAP 2022 Book 1 • Cardiology 180 Non-Statin Therapy for Dyslipidemia


Cardiology IV
Cardiology IV Panel

Series Editors: Jessica A. Bente, Pharm.D., BCPS, BCGP


Cynthia A. Sanoski, Pharm.D., FCCP, BCPS Geriatric Clinical Pharmacy Specialist
PGY-2 Geriatric Pharmacy Residency Program Director
Department Chair
Department of Pharmacy
Associate Professor of Pharmacy Practice
Cooperman Barnabas Medical Center
Thomas Jefferson University
Livingston, New Jersey
Philadelphia, Pennsylvania
Eman Elsayed Younis, Pharm.D., BCPS-AQ Cardiology
Daniel M. Witt, Pharm.D., FCCP, BCPS
Senior Clinical Pharmacist
Professor and Chair
Department of Pharmacy
Department of Pharmacotherapy
Saudi German Hospital Madinah
Assistant Dean of Clinical Affairs
Madinah, Saudi Arabia
University of Utah College of Pharmacy
Salt Lake City, Utah

Faculty Panel Chair:


Interactive Case: Anticoagulation in
Special Populations
Craig J. Beavers, Pharm.D., FCCP, FACC, FAHA, BCCP,
BCPS-AQ Cardiology, CACP Authors
Director of Cardiovascular Services Allison E. Burnett, Pharm.D., CACP
Baptist Health Paducah Antithrombosis Stewardship Pharmacist
Cardiovascular Clinical Pharmacist Inpatient Pharmacy
Department of Pharmacy Services University of New Mexico Hospital
UK Healthcare Adjunct Associate Professor of Clinical Pharmacy
Assistant Adjunct Professor President, Anticoagulation Forum
Department of Pharmacy Practice and Science University of New Mexico College of Pharmacy
University of Kentucky College of Pharmacy Albuquerque, New Mexico
Paducah, Kentucky
Kelly M. Rudd, Pharm.D., FCCP, BCPS, CACP
Clinical Associate Professor of Medicine
Interactive Case: Hyperlipidemia Department of Medical Education
Oklahoma State University Center for Health Sciences
Management for Special Populations
College of Osteopathic Medicine
Authors Tulsa, Oklahoma
Glenn Herrington, Pharm.D., AACC, BCCP, BCPS, CDCES
Reviewers
Clinical Pharmacist Practitioner
Cape Fear Heart Associates Sara R. Vazquez, Pharm.D., BCPS, CACP
Novant Health New Hanover Regional Medical Center Clinical Pharmacist
Wilmington, North Carolina Department of Pharmacy Services
Daniel M. Riche, Pharm.D., FCCP, CLS, ASH-CHC University of Utah Health
Adjunct Associate Professor in Pharmacotherapy
Professor
University of Utah College of Pharmacy
Department of Pharmacy Practice
Salt Lake City, Utah
University of Mississippi School of Pharmacy
Jackson, Mississippi Nancy Nix, Pharm.D., BCPS, BCOP
Oncology Clinical Coordinator
Reviewers Corporate Pharmacy
Elisabeth M. Wang, Pharm.D., BCCP Ballad Health
Clinical Assistant Professor Johnson City, Tennessee
Department of Pharmacy Practice and Translational
Research
University of Houston College of Pharmacy
Houston, Texas
Interactive Case: Cardiovascular Reviewers

Diseases in Pregnancy Jessie Dunne, Pharm.D., BCPS, BCCP


Clinical Pharmacy Specialist – Advanced
Author
Heart Failure and Transplant
Lindsey Federle, Pharm.D., BCCP, BCPS Department of Pharmacy
Clinical Pharmacy Specialist, Cardiology Oregon Health and Science University
Inpatient Pharmacy Portland, Oregon
UC Health – University of Cincinnati Medical Center Leslie Wooten, Pharm.D., BCPS
Cincinnati, Ohio
Clinical Manager Internal Medicine
Department of Pharmacy
AdventHealth Orlando
Orlando, Florida
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
Consultancies: Anastasia Armbruster (ACC/AHA Joint Committee Clinical Practice Guidelines); Jessica Bente (ACCP); Allison
Burnett: Consultancies (Anticoagulation Forum; National Certification Board for Anticoagulation Providers; Global Strategies);
Jessie L. Dunne (Pulmonary Hypertension Association); Nancy M. Nix (Pharmacosmos); Daniel M. Riche (Novo Nordisk; Merck;
AstraZeneca); Ralph J. Riello III (AstraZeneca; Johnson & Johnson); Sara Vazquez (Anticoagulation Forum);

Stock Ownership:

Royalties: Allison Burnett (Wolters-Kluwer); Daniel M. Riche (McGraw-Hill); Honoraria (Merck); Sara Vazquez (UpToDate)

Grants: Anastasia Armbruster (ACCP); Jessica Bente (New Jersey Society of Health-System Pharmacists); Gregory Castelli
(American Board of Family Medicine); Kevin Cowart (University of South Florida Taneja College of Pharmacy); Ralph J. Riello
III (AstraZeneca)

Honoraria: Anastasia Armbruster (AstraZeneca); John Bucheit (Association of Diabetes Care & Education Specialists); Nancy M.
Nix (Coherus; Pfizer; AstraZeneca); Ralph J. Riello III (Alexion; AstraZeneca; Janssen)

Other:

Nothing to disclose: Katherine Aymond, Eugene N. Bush, Nicholas W. Carris, Stephanie Dwyer Kaluzna, Lindsey Federle, Beth-
any A. Ford, Stormi E. Gale, Jaclynne R. Gowen, Glenn Herrington, Christine Ji, Amy L. Lehnert, Nilam R. Naik, Brenda Pahl, Joel
J. Peterson, A. Joshua Roberts, Kelly M. Rudd, Ashley Schenk, Susan M. Smith, Elisabeth M. Wang, Toby C. Trujillo, Barbara Wig-
gins, Leslie Wooten, Eman El Sayed Younis

ROLE OF BPS: The Board of Pharmacy Specialties (BPS) is an autonomous division of the American Pharmacists Association (APhA).
To maintain its strict, independent standards for certification, BPS does NOT endorse or provide review information, preparatory
courses, or study guides for Board Certification Examinations. The Board, through its specialty councils, is responsible for spe-
cialty examination content, administration, scoring, and all other aspects of its certification programs. BPS is totally separate
and distinct from ACCP. PSAP has been approved by BPS for use in BCPS recertification. Information about the BPS recertifica-
tion process is available online.

Questions regarding BPS specialty recertification should be directed to:

Board of Pharmacy Specialties


2215 Constitution Avenue NW
Washington, DC 20037
(202) 429-7591
Continuing Pharmacy Education
and Recertification Instructions
Continuing Pharmacy Education (CPE) Credit: The American College of Clinical Pharmacy is accredited by the
Accreditation Council for Pharmacy Education (ACPE) as a provider of CPE.

PSAP Target Audience: The target audience for PSAP 2022 Book 1 (Cardiology) is pharmacotherapy specialists and advanced
level clinical pharmacists encountering diverse cardiovascular patient populations.

Available CPE credits: Purchasers who successfully complete all posttests for PSAP 2022 Book 1 (Cardiology) can earn 21.5 contact
hours of CPE credit. The universal activity numbers are as follows: Cardiology I – 0217-0000-22-002-H01-P, 5.0 contact hours;
Cardiology II – 0217-0000-22-003-H01-P, 6.0 contact hours; Cardiology III – 0217-0000-22-004-H01-P, 5.0 contact hours; and
Cardiology IV – 0217-0000-22-005-H01-P, 5.5 contact hours. You may complete one or all available modules for credit. Tests may
not be submitted more than one time.

TO EARN CPE CREDITS FROM THIS PSAP BOOK

Posttest access: Go to www.accp.com and sign in with your e-mail address and password. Technical support is available from 8 a.m.
to 5 p.m. (Central) weekdays by calling (913) 492-3311. PSAP products are listed under My Products on your My Account page.

BCPS test deadline: 11:59 p.m. (Central) on July 15, 2022.


ACPE test deadline: 11:59 p.m. (Central) on January 18, 2025.

BCPS Recertification Credit: To receive BCPS recertification CPE credit, a PSAP posttest must be submitted within
the 6-month period after the book’s release (see above). Only completed tests are eligible for credit; no partial
or incomplete tests will be processed. You may complete one or all available modules for credit. Tests may not be
submitted more than one time.

The passing point to earn BCPS recertification credit is based on an expert analysis of the assessment items in each posttest
module. Any posttest submitted before the BCPS test deadline that meets this passing point will earn BCPS recertification
credits. These credits will be assigned as of the date of test submission and reported within 48 hours to BPS. For statements
of recertification credit, visit www.bpsweb.org.

Questions regarding the number of hours required for BCPS recertification should be directed to BPS at (202) 429-7591 or
www.bpsweb.org. The ACCP Recertification Dashboard is a free online tool that can track recertification credits as they are
earned through ACCP and schedule new opportunities for credits from upcoming ACCP professional development programs.

ACPE CPE Credit: To receive ACPE CPE credit for a PSAP module, a posttest must be submitted within 3 years after the book’s
release (see above). Only completed tests are eligible for credit; no partial or incomplete tests will be processed. You may
complete one or all available modules for credit. Tests may not be submitted more than one time.

Any posttest submitted before the ACPE deadline that scores 50% or greater will be awarded the appropriate CPE. These
credits will be assigned as of the date of test submission and reported within 48 hours. For statements of CPE credit, visit
www.mycpemonitor.net.

Posttest answers: The explained answers—with rationale and supporting references—will be posted s weeks after the BCPS
test deadline and will be available to anyone who has either (1) submitted a posttest or (2) waived the right to receive credit from
a posttest (see below). Go to www.accp.com and sign in with your e-mail address and password. Click the PSAP book on your
My Account page and you will see a link to the explained answers.

Test Waivers: To access the explained answers without submitting a posttest, sign in to your My Account page, select the PSAP
book, and click on the waiver link for that module. By completing the waiver form for a module, you waive the opportunity to
receive CPE credit for that module. After you submit a waiver, you will see a link to the PDF file that contains the answers for the
module you waived. Answers will be available starting 2 weeks after the BCPS test deadline.
Interactive Case: Hyperlipidemia
Management for Special Populations
By Glenn Herrington, Pharm.D., AACC, BCCP, BCPS, CDCES; and Daniel M. Riche, Pharm.D.,
FCCP, CLS, ASH-CHC

Reviewed by Elisabeth M. Wang, Pharm.D., BCCP; Jessica A. Bente, Pharm.D., BCPS, BCGP; and Eman Elsayed Younis, Pharm.D., BCPS-AQ
Cardiology

LEARNING OBJECTIVES

1. Evaluate cardiovascular risk and complications to apply pharmacologic management strategies for hyperlipidemia in
older adult patients.
2. Apply pharmacologic management strategies for hyperlipidemia in patients of various racial/ethnic backgrounds.
3. Apply pharmacologic management strategies for hyperlipidemia in patients with HIV infection.
4. Apply pharmacologic management strategies for familial hyperlipidemia.

ABBREVIATIONS IN THIS FEATURE


ASCVD Atherosclerotic cardiovascular BASELINE KNOWLEDGE STATEMENTS
disease
Readers of this feature are presumed to be familiar with the
CAC Coronary artery calcium
following:
CVD Cardiovascular disease
FH Familial hypercholesterolemia
• General knowledge of the pathophysiology of lipid
disorders
HeFH Heterozygous familial
hypercholesterolemia
• Goal-oriented, drug-oriented, and evidence-oriented strate-
gies for the treatment of lipid disorders
HoFH Homozygous familial
hypercholesterolemia • Drug knowledge of the oral and injectable pharmacologic
agents used to treat lipid disorders
PCE Pooled cohort equations
PCSK9 Proprotein convertase subtilisin/
• Statin dosing and pharmacokinetic and pharmacodynamic
parameters
kexin type 9
PLWH People living with HIV • Basic understanding of the impact of age, race, sex, genet-
ics, and concomitant diseases on lipid disorders
STEMI ST-segment elevation myocardial
infarction Table of common laboratory reference values.
T2DM Type 2 diabetes mellitus

Table of other common abbreviations. ADDITIONAL READINGS

The following free resources have additional background infor-


mation on this topic:
• Wilson PWF, Polonsky TS, Miedema MD, et al. Systematic
review for the 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/
ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the man-
agement of blood cholesterol: a report of the American
College of Cardiology/American Heart Association Task
Force on clinical practice guidelines. Circulation 2019;
139:e1144-61.

PSAP 2022 Book 1 • Cardiology 187 Interactive Case: Hyperlipidemia Management


Practice Points
• Chastain DB, Stover KR, Riche DM. Evidence-based
review of statin use in patients with HIV on antiretro- • Cardiovascular disease (CVD) is the leading cause of
viral therapy. J Clin Transl Endocrinol 2017;8:6-14. mortality worldwide. Almost 18 million people die of CVD
every year, according to the WHO. Eighty-five percent of
• Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ these deaths are a result of atherosclerotic cardiovascu-
ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/
lar disease (ASCVD), including myocardial infarction and
ASPC/NLA/PCNA guideline on the management of
stroke. Over 18 million U.S. adults have coronary artery
blood cholesterol: executive summary: a report of the
disease, according to the CDC. In addition, each year,
American College of Cardiology/American Heart
655,000 U.S. adults die of heart disease, accounting for
Association task force on clinical practice guidelines
one in four deaths.
[published correction appears in J Am Coll Cardiol
• Atherogenic lipoproteins are thought to be a strong
2019;73:3234-7]. J Am Coll Cardiol 2019;73:3168-209.
contributor to the development of ASCVD. Lipid-lowering
• Sarkar S, Brown TT. Lipid Disorders in People with drugs, primarily statins, have shown efficacy in reducing
HIV. In: Feingold KR, Anawalt B, Boyce A, et al., eds. the risk of ASCVD; however, despite strong recommenda-
Endotext [Internet]. South Dartmouth (MA): MDText. tions from several professional societies, many people at
com, Inc.; 2000. Table 5. Interaction of Antiretroviral risk of death from ASCVD are not receiving appropriate
Therapy and Statins. medication therapy. Statin-associated adverse effects, lack
• McGowan MP, Hosseini Dehkordi SH, Moriarty PM, of response, or insufficient response to maximally tolerat-
ed statins are common treatment barriers. Patient-specific
et al. Diagnosis and treatment of heterozygous
familial hypercholesterolemia. J Am Heart Assoc characteristics, such as age, race/ethnicity, sex, genetics,
2019;8:e013225. and concomitant disease states (e.g., HIV), may increase
a patient’s risk of ASCVD and affect the risk-benefit of
• Virani SS, Morris PB, Agarwala A, et al. 2021 ACC lipid-lowering drugs. Clinical pharmacists should be aware
expert consensus decision pathway on the manage- of the characteristics and risk assessment (e.g., ASCVD
ment of ASCVD risk reduction in patients with persistent risk calculator) in special populations in order to guide
hypertriglyceridemia: a report of the American Col- medication therapy recommendations and improve patient
lege of Cardiology solution set oversight committee. outcomes.
J Am Coll Cardiol 2021;78:960-93.
• Goldberg AC, Hopkins PN, Toth PP, et al. Familial
hypercholesterolemia: screening, diagnosis and man-
agement of pediatric and adult patients: clinical guid- Bohula EA, Morrow DA, Giugliano RP, et al. Atherothrombotic
ance from the National Lipid Association expert risk stratification and ezetimibe for secondary prevention.
panel on familial hypercholesterolemia. J Clin Lipidol J Am Coll Cardiol 2017;69:911–21.
2011;5:S1-8.
Carnethon MR, Pu J, Howard G, et al. Cardiovascular
health in African Americans: a scientific statement
from the American Heart Association. Circulation
2017;136:e393–423.

Conomos MP, Laurie CA, Stilp AM, et al. Genetic diversity


INTERACTIVE CASE: and association studies in US Hispanic/Latino popula-
HYPERLIPIDEMIA MANAGEMENT tions: applications in the Hispanic Community Health
FOR SPECIAL POPULATIONS Study/Study of Latinos. Am J Hum Genet 2016;98:165–84.
• Click here to start this PSAP activity. Currier JS, Lundgren JD, Carr A, et al. Epidemiological evi-
dence for cardiovascular disease in HIV-infected patients
REFERENCES and relationship to highly active antiretroviral therapy.
Circulation 2008;118:e29–e35.
Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA
guideline on the primary prevention of cardiovascular Daviglus ML, Pirzada A, Talavera GA. Cardiovascular disease
disease: a report of the American College of Cardiology/ risk factors in the Hispanic/Latino population: lessons
American Heart Association Task Force on Clinical from the Hispanic Community Health Study/Study of
Practice Guidelines. J Am Coll Cardiol 2019;74:e177-e232. Latinos (HCHS/SOL). Prog Cardiovasc Dis 2014;57:230–6.
Baigent, C, Blackwell, L, Emberson, J, et al. Efficacy and Daviglus ML, Pirzada A, Durazo-Arvizu R, et al. Prevalence
safety of more intensive lowering of LDL cholesterol: of low cardiovascular risk profile among diverse Hispanic/
a meta-analysis of data from 170 000 participants in Latino adults in the United States by age, sex, and level
26 randomised trials. Lancet. 2010;376:1670–81 of acculturation: the Hispanic Community Health Study/
Study of Latinos. J Am Heart Assoc 2016;5:e003929.
Birmingham BK, Bujac SR, Elsby R, et al. Rosuvastatin
pharmacokinetics and pharmacogenetics in white and Ford ES, Li C, Zhao G. Prevalence and correlates of metabolic
Asian subjects residing in the United States. Eur J Clin syndrome based on a harmonious definition among adults
Pharmacol 2015;71:329–40. in the US. J Diabetes 2010;2:180–93.

PSAP 2022 Book 1 • Cardiology 188 Interactive Case: Hyperlipidemia Management


Frank, AT, Zhao, B, Jose, PO, et al. Racial/ethnic differences Mortensen MB, Falk E. Primary prevention with statins in the
in dyslipidemia patterns. Circulation. 2014;129:570–9. elderly. J Am Coll Cardiol 2018;71:85–94.

Garfinkel D, Ilhan B, Bahat G. Routine deprescribing of Muntner P, Colantonio LD, Cushman M, et al. Validation of
chronic medications to combat polypharmacy. Ther Adv the atherosclerotic cardiovascular disease Pooled Cohort
Drug Saf 2015;6:212–33. risk equations. JAMA 2014;311:1406–15.

Gnjidic D, Le Couteur DG, Blyth FM, et al. Statin use Pejic RN. Familial hypercholesterolemia. Ochsner J 2014
and clinical outcomes in older men: a prospective Winter;14:669-72.
population-based study. BMJ Open 2013;3:e002333.
Pilotto A, Panza F, Copetti M, et al. Statin treatment and
Goff DC Jr, Lloyd-Jones DM, Bennett G, et al. 2013 ACC/ mortality in community-dwelling frail older patients
AHA guideline on the assessment of cardiovascular with diabetes mellitus: a retrospective observational
risk: a report of the American College of Cardiology/ study. PLoS One 2015:10;e0130946.
American Heart Association Task Force on Practice
Guidelines. Circulation 2014;129:S49–73. Pu J, Romanelli R, Zhao B, et al. Dyslipidemia in special
ethnic populations. Cardiol Clin 2015;33:325–33.
Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/
AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/ Qi K, Reeve E, Hilmer SN, et al. Older peoples’ attitudes
NLA/PCNA guideline on the management of blood regarding polypharmacy, statin use and willingness to
cholesterol: executive summary: a report of the American have statins deprescribed in Australia. Int J Clin Pharm
College of Cardiology/American Heart Association 2015;37:949–57.
Task Force on Clinical Practice Guidelines. J Am Coll Qureshi WT, Kaplan RC, Swett K, et al. American College of
Cardiol 2019;73:3168-209. Cardiology/American Heart Association (ACC/AHA) class
Gujral UP, Vittinghoff E, Mongraw-Chaffin M, et al. Car- I guidelines for the treatment of cholesterol to reduce
diometabolic abnormalities among normal-weight persons atherosclerotic cardiovascular risk: implications for US
from five racial/ethnic groups in the United States: a Hispanics/Latinos based on findings from the Hispanic
cross-sectional analysis of two cohort studies. Ann Intern Community Health Study/Study of Latinos (HCHS/SOL).
Med 2017;166:628–36. J Am Heart Assoc 2017:6;e005045.

Hata J, Kiyohara Y. Epidemiology of stroke and coronary Rana JS, Tabada GH, Solomon MD, et al. Accuracy of the
artery disease in Asia. Circ J 2013;77:1923–32. atherosclerotic cardiovascular risk equation in a large
contemporary, multiethnic population. J Am Coll Cardiol
Hutchinson RN, Shin S. Systematic review of health 2016;67:2118–30.
disparities for cardiovascular diseases and associ-
ated factors among American Indian and Alaska Native Rao G, Powell-Wiley TM, Ancheta I, et al. Identification of
populations. PLoS One 2014:9;e80973. obesity and cardiovascular risk in ethnically and racially
diverse populations: a scientific statement from the
Kutner JS, Blatchford PJ, Taylor DH Jr, et al. Safety and American Heart Association. Circulation 2015;132:457–72.
benefit of discontinuing statin therapy in the setting
of advanced, life-limiting illness: a randomized clinical Rasheed S, Yan JS, Lau A, Chan AS. HIV replication
trial. JAMA Intern Med 2015;175:691–700. enhances production of free fatty acids, low density
lipoproteins and many key proteins involved in lipid
Lambert CT, Sandesara P, Isiadinso I, et al. Current metabolism: a proteomics study, PLoS ONE 2008;3:e3003.
treatment of familial hypercholesterolaemia. Eur Cardiol
2014;9:76–81. Riddler SA, Smit E, Cole SR, et al. Impact of HIV infection and
HAART on serum lipids in men. JAMA 2003;289:2978–82.
Liao JK. Safety and efficacy of statins in Asians. Am J
Cardiol 2007;99:410–4. Rodriguez F, Maron DJ, Knowles JW, et al. Association
between intensity of statin therapy and mortality in
Mahabadi AA, Mohlenkamp S, Lehmann N, et al. CAC score patients with atherosclerotic cardiovascular disease.
improves coronary and CV risk assessment above statin JAMA Cardiol 2017;2:47–54.
indication by ESC and AHA/ACC primary prevention
guidelines. JACC Cardiovasc Imaging 2017;10:143–53. Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab
and clinical outcomes in patients with cardiovascular
McGowan MP, Hosseini Dehkordi SH, Moriarty PM, Duell PB. disease. N Engl J Med 2017;376:1713–22.
Diagnosis and treatment of heterozygous familial hyper-
cholesterolemia. J Am Heart Assoc 2019;8(24):e013225. Savarese G, Gotto AM Jr, Paolillo, S, et al. Benefits of statins
in elderly subjects without established cardiovascular
Molina JM, Andrade-Villanueva JEchevarria J, et al. Once- disease: a meta-analysis. J Am Coll Cardiol
daily atazanavir/ritonavir compared with twice-daily 2013;62:2090–9.
lopinavir/ritonavir, each in combination with tenofovir
and emtricitabine, for management of antiretroviral-naïve Schwartz GG, Steg PG, Szarek M, et al. Alirocumab and
HIV-1-infected patients: 96-week efficacy and safety cardiovascular outcomes after acute coronary syndrome.
results of the CASTLE study. J Acquir Immune Defic Syndr N Engl J Med 2018;379:2097-107.
2010;53:323–32.

PSAP 2022 Book 1 • Cardiology 189 Interactive Case: Hyperlipidemia Management


Shepherd J, Blauw GJ, Murphy MB, et al. Pravastatin in Volgman AS, Palaniappan LS, Aggarwal NT, et al. Atheroscle-
elderly individuals at risk of vascular disease (PROSPER): rotic cardiovascular disease in South Asians in the United
a randomised controlled trial. Lancet 2002;360:1623–30. States: epidemiology, risk factors, and treatments: a
scientific statement from the American Heart Association.
Tebas P, Henry WK, Matining R, et al. Metabolic and immune Circulation 2018;138:e1–34.
activation effects of treatment interruption in chronic
HIV-1 infection: implications for cardiovascular risk. PLoS Wilson PWF, Polonsky TS, Miedema MD, et al. System-
ONE 2008;3:e2021. atic review for the 2018 AHA/ACC/AACVPR/AAPA/ABC/
ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the
Teng M, Lin L, Zhao YJ, et al. Statins for primary prevention management of blood cholesterol: a report of the Amer-
of cardiovascular disease in elderly patients: systematic ican College of Cardiology/American Heart Association
review and meta-analysis. Drugs Aging 2015;32:649–61. Task Force on Clinical Practice Guidelines. Circulation
Thai M, Reeve E, Hilmer SN, et al. Prevalence of statin-drug 2019;139:e1144–61. 
interactions in older people: a systematic review. Eur J Writing Committee, Virani SS, Morris PB, Agarwala A, Bal-
Clin Pharmacol 2016;72:513–21. lantyne CM, et al. 2021 ACC Expert Consensus Decision
Tikkanen, MJ, Holme, I, Cater, NB, et al. Comparison of Pathway on the Management of ASCVD Risk Reduction
efficacy and safety of atorvastatin (80 mg) to simvastatin in Patients With Persistent Hypertriglyceridemia:
(20 to 40 mg) in patients aged <65 versus >65 years with A Report of the American College of Cardiology Solution
coronary heart disease (from the Incremental DEcrease Set Oversight Committee. J Am Coll Cardiol. 2021 Jul
through Aggressive Lipid Lowering [IDEAL] study) Am J 22:S0735-1097(21)05323-7.
Cardiol. 2009;103:577–82

PSAP 2022 Book 1 • Cardiology 190 Interactive Case: Hyperlipidemia Management


Self-Assessment Questions
Questions 1 and 2 pertain to the following case. C. Ezetimibe may be preferred to a statin because of its
M.J., an 83-year-old white woman with type 2 diabetes melli- lower risk of adverse effects.
tus (T2DM) and hypertension, is referred to a pharmacist-led D. Statin therapy should be avoided because of the
lipid clinic for management of hyperlipidemia. The referral potential for worsening control of diabetes.
from her primary care provider indicates that her LDL choles-
4. Which one of the following is the best goal LDL choles-
terol is “not at goal” and includes previous adverse drug reac-
terol to recommend for S.M.?
tions to atorvastatin, rosuvastatin, and simvastatin. M.J.’s
most recent lipid panel shows TC 205 mg/dL, LDL cholesterol A. Cannot determine because of his age
124 mg/dL, HDL cholesterol 48 mg/dL, and TG 88 mg/dL. Her B. Less than 70 mg/dL
A1C is 6.1% and blood pressure is 118/68 mm Hg. M.J.’s only C. Less than 100 mg/dL
current medication is pitavastatin 1 mg daily. She admits that D. 50% reduction from baseline
she often skips doses because of myalgias that limit her abil- 5. A 53-year-old man of South Asian ancestry has a medical
ity to work in her garden. She refuses to try another statin history of hypertension (currently treated with amlodip-
because of severe myalgias with previous statin therapy. M.J. ine 5 mg once daily). His blood pressure today is 118/68
would prefer to manage her atherosclerotic cardiovascular mm Hg. A lipid panel includes TC 185 mg/dL, HDL cho-
disease (ASCVD) risk with diet and exercise. lesterol 30 mg/dL, LDL cholesterol 130 mg/dL, and TG
1. Which one of the following best assesses M.J.’s 10-year 128 mg/dL. The patient is not a smoker. He asks about
ASCVD risk? lipid-lowering medications because his father died of a
MI at 53 years of age. According to the ACC/AHA pooled
A. Less than 7.5%
cohort equations (PCE), the patient’s 10-year ASCVD risk
B. 7.5% to less than 20%
is 6.9%. Which one of the following is best to recommend
C. At least 20%
for managing this patient’s blood cholesterol?
D. Cannot estimate because of her age
A. No medication changes are indicated.
2. Which one of the following is best to recommend for
B. Initiate rosuvastatin 5 mg daily.
management of M.J.’s hyperlipidemia?
C. Initiate rosuvastatin 20 mg daily.
A. Discontinue pitavastatin. D. Initiate evolocumab 140 mg every 14 days.
B. Continue pitavastatin and add ezetimibe.
6. A 65-year-old Japanese American woman has a medical
C. Discontinue pitavastatin and initiate pravastatin
history that includes hypertension and T2DM. Simvasta-
10 mg once daily.
tin 20 mg once daily was initiated 2 weeks ago. Today,
D. Discontinue pitavastatin and initiate ezetimibe.
she presents to the ED with new-onset, severe bilateral
cramping of the deltoids. Her CK concentration is within
Questions 3 and 4 pertain to the following case.
normal limits. Which one of the following is best to rec-
S.M., a 76-year-old African American man, is seen for ommend regarding this patient’s myalgia?
a 1-month follow-up after hospitalization for an acute
A. Continue simvastatin.
ST-segment elevation myocardial infarction (STEMI). His
B. Change simvastatin to atorvastatin 20 mg once daily.
medical history includes T2DM and hypertension. S.M.’s LDL
C. Change simvastatin to rosuvastatin 20 mg once daily.
cholesterol at the time of the STEMI was 155 mg/dL. Rosu-
D. Change simvastatin to ezetimibe.
vastatin 40 mg daily was initiated during his hospitalization.
7. A 51-year-old man moved to the mainland United States
3. S.M. has never taken a lipid-lowering agent and voices
from Puerto Rico after losing his home in a tropical
concerns about newly prescribed rosuvastatin after
storm. He is referred to cardiology because of a recent
hearing of potential adverse drug effects from his neigh-
diagnosis of severe primary hypercholesterolemia with
bors. Which one of the following is the best educational
coronary artery disease. The patient has taken rosuvas-
point to share with S.M. regarding rosuvastatin?
tatin 40 mg once daily for several years. His lipid panel
A. Statin therapy is recommended because of his very today includes TC 247 mg/dL, HDL cholesterol 41 mg/dL,
high risk of another ASCVD event. LDL cholesterol 198 mg/dL, and TG 105 mg/dL. The car-
B. Statin therapy is not recommended because of lack diologist wants to initiate a proprotein convertase sub-
of evidence in patients older than 75. tilisin/kexin type 9 (PCSK9) inhibitor; however, there is
concern about its cost because the patient is unem-
ployed and has no prescription insurance. Which one

PSAP 2022 Book 1 • Cardiology 191 Interactive Case: Hyperlipidemia Management


of the following is best to recommend for this patient’s 1 mg subcutaneously once weekly, olmesartan 40 mg
hypercholesterolemia? once daily, rosuvastatin 40 mg once daily, ezetimibe
10 mg once daily, and 4 days remaining of triple ther-
A. Provide an evolocumab manufacturer’s co-pay card.
apy with clarithromycin 500 mg twice daily, amoxicillin
B. Initiate ezetimibe rather than a PCSK9 inhibitor.
1000 mg twice daily, and pantoprazole 40 mg twice daily.
C. Change rosuvastatin 40 mg once daily to
His LDL cholesterol today is 197 mg/dL, which remains
atorvastatin 80 mg once daily.
above goal. The patient worries about remembering
D. Help the patient apply to the evolocumab
to take another medication because he says he barely
manufacturer’s patient assistance foundation.
remembers to even take his current medications. Which
one of the following is best to recommend to help further
Questions 8 and 9 pertain to the following case.
reduce this patient’s LDL cholesterol?
K.M., a 50-year-old African American man, presents to car-
diometabolic clinic for a follow-up. His medical history is A. Niacin
significant for newly diagnosed HIV infection, controlled B. Colesevelam
mitochondrial myopathy, and controlled hypertension. C. Fish oil
K.M.’s lipid panel today shows HDL cholesterol 38 mg/dL, D. Alirocumab
TG 200 mg/dL, and LDL cholesterol 188 mg/dL, consistent
with the laboratory values at his first appointment 3 months Questions 11 and 12 pertain to the following case.

ago. The patient’s medication history includes darunavir/ R.W., a 23-year-old white woman, presents to your cardiomet-
cobicistat/emtricitabine/tenofovir alafenamide (Symtuza) abolic clinic for a follow-up. Her medical history is significant
and hydrochlorothiazide. In addition, because of a low CD4 for HeFH caused by a mutation in the LDL receptor protein.
count, K.M. takes trimethoprim/sulfamethoxazole and fluco- R.W. has not had an ASCVD event. Today, R.W.’s LDL choles-
nazole for primary opportunistic infection prophylaxis. terol is 230 mg/dL and TG are 298 mg/dL. She currently takes
rosuvastatin 40 mg/day, ezetimibe 10 mg/day, and ethinyl
8. Which one of the following is best to recommend initiat-
estradiol/etonogestrel vaginal ring.
ing to help reduce K.M.’s LDL cholesterol and ASCVD risk?
11. Although she has previously discussed starting a PCSK9
A. Atorvastatin 40 mg/day
inhibitor, R.W. is still resistant because of her fear of nee-
B. Pitavastatin 2 mg/day
dles. She wants to know what other oral options are avail-
C. Pravastatin 40 mg/day
able. Which one of the following is best to recommend
D. Fluvastatin 20 mg/day
adding to R.W.’s current therapy?
9. K.M. started the recommended statin therapy. He returns
A. Bempedoic acid
to your clinic after being discharged from the hospital
B. Evolocumab
5 days ago for hypertriglyceridemia-induced acute pan-
C. Fenofibrate
creatitis. His inpatient laboratory test results included
D. Niacin
TG 401 mg/dL and LDL cholesterol 172 mg/dL. None
of K.M.’s medications before the hospitalization were 12. Six months later, R.W. returns to the clinic for a follow-up.
changed at discharge, and he was referred back to your Her LDL cholesterol has improved to 198 mg/dL but TG
clinic for outpatient follow-up and medication manage- are 304 mg/dL. She announces that she recently became
ment. Which one of the following is best to recommend pregnant (currently second trimester). R.W. takes rosu-
for K.M. regarding statin? vastatin 40 mg/day, ezetimibe 10 mg/day, colesevelam
3.75 g/day, and a prenatal vitamin. After counseling,
A. Change to atorvastatin 40 mg/day and add fish oil.
R.W. wants to prioritize as little harm as possible to the
B. Change to rosuvastatin 20 mg/day and add
pregnancy. In addition to incorporating lifestyle changes
fenofibrate.
focused on maintaining a diet low in fats and cholesterol,
C. Change to simvastatin 20 mg/day and add
which one of the following is best to recommend for R.W.?
ezetimibe.
D. Change to rosuvastatin 20 mg/day and add A. Continue current regimen; add evolocumab for
gemfibrozil. further LDL cholesterol lowering.
B. Discontinue rosuvastatin; continue ezetimibe and
10. A 42-year-old African American man presents to the
colesevelam.
clinic for his 3-month follow-up. His medical history is
C. Discontinue rosuvastatin and ezetimibe; continue
significant for heterozygous familial hypercholesterol-
colesevelam.
emia (HeFH), T2DM, and a recent hospital admission for
D. Discontinue rosuvastatin, ezetimibe, and
a Helicobacter pylori infection. The patient’s home drugs
colesevelam; initiate lipoprotein apheresis.
include metformin 1000 mg twice daily, semaglutide

PSAP 2022 Book 1 • Cardiology 192 Interactive Case: Hyperlipidemia Management


Learner Feature Evaluation: Interactive Case: Hyperlipidemia Management
for Special Populations

As you take the posttest for this feature, also evaluate the 7. The content of the feature was useful to me.
material’s quality and usefulness, as well as the achieve- 8. The teaching and learning methods used in the feature
ment of learning objectives. Rate each item using this 5-point were effective.
scale: 9. The active learning methods used in the feature were
effective.
• Strongly agree
• Agree 10. The learning assessment activities used in the feature
• Neutral were effective.
• Disagree 11. The feature was effective overall.
• Strongly disagree 12. The activity met the stated learning objectives.
1. The content of the feature met my educational needs. 13. If any objectives were not met, please list them here.
2. The content of the feature satisfied my expectations.
3. The author presented the feature content effectively. OTHER COMMENTS
4. The content of the feature was relevant to my practice 14. Please provide any specific comments related to any
and presented at the appropriate depth and scope. perceptions of bias, promotion, or advertisement of
5. The content of the feature was objective and balanced. commercial products.

6. The content of the feature is free of bias, promotion, and 15. Please expand on any of your above responses, and/or
advertisement of commercial products. provide any additional comments regarding this feature:

PSAP 2022 Book 1 • Cardiology 193 Interactive Case: Hyperlipidemia Management


Interactive Case: Anticoagulation in
Special Populations
By Allison E. Burnett, Pharm.D., CACP; and Kelly M. Rudd, Pharm.D., FCCP, BCPS, CACP

Reviewed by Sara R. Vazquez, Pharm.D., BCPS, CACP; and Nancy Nix, Pharm.D., BCPS, BCOP

LEARNING OBJECTIVES

1. Evaluate available evidence pertaining to the use of oral anticoagulants in patients with antiphospholipid syndrome
(APS).
2. Apply appropriate diagnostic modalities and construct an anticoagulant therapy plan for patients with suspected or
confirmed heparin-induced thrombocytopenia.
3. Evaluate the risk-benefit of vitamin K antagonist and direct oral anticoagulant (DOAC) therapies in patients with renal
impairment.
4. Assess the role of DOAC therapy in patients at extremes of weight.

ABBREVIATIONS IN THIS FEATURE


AC Anticoagulant/anticoagulation BASELINE KNOWLEDGE STATEMENTS

aCL Anticardiolipin antibody Readers of this feature are presumed to be familiar with the
AF Atrial fibrillation following:
APLA Antiphospholipid antibody • Physiology of immune system function
APS Antiphospholipid syndrome • Normal coagulation physiology
β2GPI β2 glycoprotein I
• General knowledge of anticoagulants (ACs) and associ-
CAPS Catastrophic APS ated dosing regimens
CFX Chromogenic factor X • General knowledge of pathophysiologic conditions requir-
CKD Chronic kidney disease ing AC therapy
CRNMB Clinically relevant nonmajor • General guideline recommendations for the treatment of
bleeding conditions requiring AC therapy
DOAC Direct oral anticoagulant
Table of common laboratory reference values
DVT Deep venous thrombosis
ECMO Extracorporeal membrane
oxygenation ADDITIONAL READINGS
ELISA Enzyme-linked immunosorbent
assay The following resources have additional background informa-
ESRD End-stage renal disease tion on these topics:
HIPA Heparin-induced platelet aggrega- • EULAR recommendations for the management of
tion assay antiphospholipid syndrome in adults. Ann Rheum Dis
HIT Heparin-induced 2019;78:1296-304.
thrombocytopenia • Diagnosis and management of the antiphospholipid
HITT HIT with thrombosis syndrome. N Engl J Med 2018;378:2010-21.
IVC Inferior vena cava • Use of DOACs in obesity for treatment and prevention of
LA Lupus anticoagulant venous thromboembolism: updated communication from
the ISTH SSC Subcommittee on Control of
LAAO Left atrial appendage occlusion
Anticoagulation. J Thromb Haemost 2021;19:1874-82.
MB Major bleeding
NVAF Nonvalvular atrial fibrillation

PSAP 2022 Book 1 • Cardiology 195 Interactive Case: Anticoagulation


OAC Oral anticoagulant • Chronic kidney disease and cerebrovascular disease:
PCI Percutaneous coronary intervention consensus and guidance from a KDIGO Controver-
PE Pulmonary embolism sies Conference. Stroke 2021;52:e328-e346.
PTCP Pretest clinical probability • Oral anticoagulation for patients with atrial fibrilla-
RCT Randomized controlled trial tion on long-term dialysis. JACC 2020;75:273-85.
SLE Systemic lupus erythematosus • American Society of Hematology guideline for man-
agement of venous thromboembolism: heparin-in-
TTR Time in therapeutic INR range
duced thrombocytopenia. Blood Adv 2018;2:3360-92.
VTE Venous thromboembolism

Table of other common abbreviations.

Bansal VK, Herzog CA, Sarnak MJ, et al. Oral anticoagu-


INTERACTIVE CASE: lants to prevent stroke in nonvalvular atrial fibrillation in
patients with CKD stage 5D: an NKF-KDOQI Controversies
ANTICOAGULATION IN SPECIAL
Report. Am J Kidney Dis 2017;70:859-68.
POPULATIONS
Barlow A, Barlow B, Reinaker T, et al. Potential role of
• Click here to start this PSAP activity direct oral anticoagulants in the management of hep-
arin-induced thrombocytopenia. Pharmacotherapy
2019;39:837-53.

Boersma LV, Ince H, Kische S, et al. Evaluating real-world


Practice Points
clinical outcomes in atrial fibrillation patients receiving
• Hypercoagulable conditions such as antiphospholipid the WATCHMAN left atrial appendage closure technol-
syndrome (APS) and heparin-induced thrombocytopenia ogy: final 2-year outcome data of the EWOLUTION trial
(HIT) are associated with complex diagnostic workups and focusing on history of stroke and hemorrhage. Circulation
may require use of different management approaches from Arrhythmia and Electrophysiology 2019;12:e006841
those used for routine thromboembolic events such as
deep venous thrombosis (DVT) and pulmonary embolism Boonyawat K, Caron F, Li A, et al. Association of body
(PE). weight with efficacy and safety outcomes in phase III
• Pharmacists should have good familiarity with who should randomized controlled trials of direct oral anticoagu-
and should not be tested for APS and HIT, interpretation of lants: a systematic review and meta-analysis. J Thromb
laboratory results, and appropriate pharmacotherapeutic Haemost 2017;15:1322-33.
options specific to these conditions.
• Obese and renal impairment populations have been un- Boriani G, Ruff CT, Kuder JF, et al. Relationship between
derrepresented in the large randomized controlled trials body mass index and outcomes in patients with atrial
(RCTs) comparing direct oral anticoagulants (DOACs) with fibrillation treated with edoxaban or warfarin in the
warfarin for venous thromboembolism (VTE) and non- ENGAGE AF-TIMI 48 trial. Eur Heart J 2019;40:1541-50.
valvular atrial fibrillation (NVAF), but emerging evidence
Brodsky SV, Satoskar A, Chen J, et al. Acute kidney injury
suggests DOACs and warfarin are reasonable for treating
during warfarin therapy associated with obstructive
and preventing thrombosis in these populations.
tubular red blood cell casts: a report of 9 cases. Am J
• Pharmacists play an essential role in stewardship over
Kidney Dis 2009;54:1121-6.
high-risk antithrombotic therapies, especially in special
populations of patients who may be more vulnerable to Brouwer TF, Whang W, Kuroki K, et al. Net clinical benefit of
adverse events and poor outcomes. left atrial appendage closure versus warfarin in patients
with atrial fibrillation: a pooled analysis of the randomized
PROTECT-AF and PREVAIL studies. J Am Heart Assoc
2019;8:e013525.
REFERENCES Camm AJ, Cools F, Virdone S, et al. Mortality in patients
Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the with atrial fibrillation receiving nonrecommended
treatment of acute venous thromboembolism. N Engl J doses of direct oral anticoagulants. J Am Coll Cardiol
Med 2013;369:799-808. 2020;76:1425-36.

Ahuja T, Sessa K, Merchan C, et al. Oral factor Xa inhibitors Cervera R, Piette JC, Font J, et al. Antiphospholipid
versus warfarin for the treatment of venous thromboem- syndrome: clinical and immunologic manifestations
bolism in advanced chronic kidney disease. Adv Hematol and patterns of disease expression in a cohort of 1,000
2021;2021:e8870015. patients. Arthritis Rheum 2002;46:1019-27.

Arepally GM, Padmanabhan A. Heparin-induced thrombocy- Cervera R, Rodríguez-Pintó I, Colafrancesco S, et al. 14th
topenia: a focus on thrombosis. Arterioscler Thromb Vasc International Congress on Antiphospholipid Antibodies
Biol 2021;41:141-52.

PSAP 2022 Book 1 • Cardiology 196 Anticoagulation in Special Populations


Task Force Report on Catastrophic Antiphospholipid Dias C, Moore KT, Murphy J, et al. Pharmacokinetics, phar-
Syndrome. Autoimmun Rev 2014;13:699-707. macodynamics, and safety of single-dose rivaroxaban in
chronic hemodialysis. Am J Nephrol 2016;43:229-36.
Cervera R, Serrano R, Pons-Estel GJ, et al. Morbidity and
mortality in the antiphospholipid syndrome during a Dobesh PP, Kernan MM, Lueshen JJ. Direct oral anticoagu-
10-year period: a multicentre prospective study of 1000 lants in the treatment of venous thromboembolism: use in
patients. Ann Rheum Dis 2015;74:1011-8. patients with advanced renal impairment, obesity, or other
weight-related special populations. Semin Respir Crit Care
Chang M, Yu Z, Shenker A, et al. Effect of renal impairment Med 2021;42:233-49.
on the pharmacokinetics, pharmacodynamics, and safety
of apixaban. J Clin Pharmacol 2016;56:637-45. Doré M, Frenette A, Chagnon I, et al. Interrater agreement for
two systems used to determine the probability of hepa-
Cohen H, Efthymiou M, Isenberg DA. Use of direct oral rin-induced thrombocytopenia. Am J Health Syst Pharm
anticoagulants in antiphospholipid syndrome. J Thromb 2014;71:2045-52.
Haemost 2018;16:1028-39.
Douketis JD, Spyropoulos AC, Spencer FA, et al.
Cohen H, Isenberg DA. How I treat anticoagulant-refrac- Perioperative management of antithrombotic therapy:
tory thrombotic antiphospholipid syndrome. Blood Antithrombotic Therapy and Prevention of Thrombosis,
2021;137:299-309. 9th ed: American College of Chest Physicians
Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus Evidence-Based Clinical Practice Guidelines. Chest
warfarin in patients with atrial fibrillation. N Engl J Med 2012;141(2 suppl):e326S-e350S.
2009;361:1139-51. Dreisbach AW. The influence of chronic renal failure on
Cuker A, Arepally GM, Chong BH, et al. American Society of drug metabolism and transport. Clin Pharmacol Ther
Hematology 2018 guidelines for management of venous 2009;86:553-6.
thromboembolism: heparin-induced thrombocytopenia. EINSTEIN-PE Investigators, Büller HR, Prins MH, et al. Oral
Blood Adv 2018;2:3360-92. rivaroxaban for the treatment of symptomatic pulmonary
Cuker A, Cines DB. How I treat heparin-induced embolism. N Engl J Med 2012;366:1287-97.
thrombocytopenia. Blood 2012a;119:2209-18. Erkan D, Espinosa G, Cervera R. Catastrophic antiphospho-
Cuker A, Gimotty PA, Crowther MA, et al. Predic- lipid syndrome: updated diagnostic algorithms. Autoim-
tive value of the 4Ts scoring system for hepa- mun Rev 2010;10:74-9.
rin-induced thrombocytopenia: a systematic review and Fanikos J, Burnett AE, Mahan CE, et al. Renal function
meta-analysis. Blood 2012b;120:4160-7. considerations for stroke prevention in atrial fibrillation.
de Groot PG, Lutters B, Derksen RHWM, et al. Lupus Am J Med 2017a;130:1015-23.
anticoagulants and the risk of a first episode of deep Fanikos J, Burnett AE, Mahan CE, et al. Renal function and
venous thrombosis. J Thromb Haemost 2005;3:1993-7. direct oral anticoagulant treatment for venous thrombo-
Dentali F, Manfredi E, Crowther M, Ageno W. Long-duration embolism. Am J Med 2017b;130:1137-43.
therapy with low molecular weight heparin in patients with Gallagher D, Visser M, Sepúlveda D, et al. How useful is body
antiphospholipid antibody syndrome resistant to warfarin mass index for comparison of body fatness across age,
therapy. J Thromb Haemost. 2005 Sep;3(9):2121-3. sex, and ethnic groups? Am J Epidemiol 1996;143:228-39.
De Vriese AD, Caluwé R, Meersch HVD, et al. Safety and Galli M, Luciani D, Bertolini G, et al. Lupus anticoagulants are
efficacy of vitamin K antagonists versus rivaroxaban stronger risk factors for thrombosis than anticardiolipin
in hemodialysis patients with atrial fibrillation: a antibodies in the antiphospholipid syndrome: a systematic
multicenter randomized controlled trial. J Am Soc Nephrol review of the literature. Blood 2003;101:1827-32.
2021;32:1474-83.
Garcia D, Erkan D. Diagnosis and management of the anti-
De Vriese AS, Caluwé R, Bailleul E, et al. Dose-finding study phospholipid syndrome. N Engl J Med 2018;378:2010-21.
of rivaroxaban in hemodialysis patients. Am J Kidney Dis
2015;66:91-8. Garza-Mayers AC, Shah R, Sykes DB, et al. The successful
use of apixaban in dialysis patients with calciphylaxis who
Devreese KMJ, de Groot PG, de Laat B, et al. Guidance from require anticoagulation: a retrospective analysis. Am J
the Scientific and Standardization Committee for lupus Nephrol 2018;48:168-71.
anticoagulant/antiphospholipid antibodies of the Interna-
tional Society on Thrombosis and Haemostasis. J Thromb Gebhart J, Posch F, Koder S, et al. Increased mortality in
Haemost 2020;18:2828-39. patients with the lupus anticoagulant: the Vienna Lupus
Anticoagulant and Thrombosis Study (LATS). Blood
Di Minno MND, Lupoli R, Di Minno A, et al. Effect of 2015;125:3477-83.
body weight on efficacy and safety of direct oral
anticoagulants in the treatment of patients with acute Ghoshal S, Freedman BI. Mechanisms of stroke in
venous thromboembolism: a meta-analysis of randomized patients with chronic kidney disease. Am J Nephrol
controlled trials. Ann Med 2015;47:61-8. 2019;50:229-39.

PSAP 2022 Book 1 • Cardiology 197 Anticoagulation in Special Populations


Goto S, Angchaisuksiri P, Bassand J, et al. Management and Kidney Disease: Improving Global Outcomes (KDIGO). CKD
1-year outcomes of patients with newly diagnosed atrial Evaluation and Management. 2021.
fibrillation and chronic kidney disease: results from the
prospective GARFIELD-AF Registry. J Am Heart Assoc Kooiman J, van der Hulle T, Maas H, et al. Pharmacokinet-
2019;8:e010510. ics and pharmacodynamics of dabigatran 75 mg b.i.d. in
patients with severe chronic kidney disease. J Am Coll
Granger CB, Alexander JH, McMurray JJV, et al. Apixaban Cardiol 2016;67:2442-4.
versus warfarin in patients with atrial fibrillation. N Engl
J Med 2011;365:981-92. Krnic-Barrie S, O’Connor CR, Looney SW, et al. A retrospec-
tive review of 61 patients with antiphospholipid syndrome.
Greinacher A. Heparin-induced thrombocytopenia. N Engl Analysis of factors influencing recurrent thrombosis. Arch
J Med 2015;373:252-61. Intern Med 1997;157:2101-8.

Gulseth MP, Wittkowsky AK, Fanikos J, et al. Safety and Kubitza D, Becka M, Mueck W, et al. Effects of renal impair-
efficacy of vitamin K antagonists versus rivaroxaban ment on the pharmacokinetics, pharmacodynamics and
in hemodialysis patients with atrial fibrillation: a multi- safety of rivaroxaban, an oral, direct factor Xa inhibitor.
center randomized controlled trial. Pharmacotherapy Br J Clin Pharmacol 2010;70:703-12.
2011;31:1232-49.
Kubitza D, Becka M, Zuehlsdorf M, et al. Body weight has
Hanni C, Petrovitch E, Ali M, et al. Outcomes associated with limited influence on the safety, tolerability, pharmacokinet-
apixaban vs warfarin in patients with renal dysfunction. ics, or pharmacodynamics of rivaroxaban (BAY 59-7939) in
Blood Adv 2020;4:2366-71. healthy subjects. J Clin Pharmacol 2007;47:218-26.

Herndon K, Guidry TJ, Wassell K, et al. Characterizing the Kuno T, Takagi H, Ando T, et al. Oral anticoagulation for
safety profile of apixaban versus warfarin in moderate to patients with atrial fibrillation on long-term dialysis. J Am
severe chronic kidney disease at a Veterans Affairs hospi- Coll Cardiol 2020;75:273-85.
tal. Ann Pharmacother 2020;54:554-60.
Liesenfeld KH, Lehr T, Dansirikul C, et al. Population pharma-
Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines cokinetic analysis of the oral thrombin inhibitor dabiga-
for the diagnosis and management of atrial fibrillation tran etexilate in patients with non-valvular atrial fibrillation
developed in collaboration with the European Association from the RE-LY trial. J Thromb Haemost 2011;9:2168-75.
for Cardio-Thoracic Surgery (EACTS): the task force for
the diagnosis and management of atrial fibrillation of the Limdi NA, Beasley TM, Baird MF, et al. Kidney function influ-
European Society of Cardiology (ESC) developed with the ences warfarin responsiveness and hemorrhagic compli-
special contribution of the European Heart Rhythm Asso- cations. J Am Soc Nephrol 2009;20:912-21.
ciation (EHRA) of the ESC. Eur Heart J 2021;42:373-498. Linkins LA, Dans AL, Moores LK, et al. Treatment and preven-
Htet S, Hayes L, Leung T. Strong lupus anticoagulant (LA) tion of heparin-induced thrombocytopenia: Antithrombotic
as a cause for prolonged prothrombin time (PT), activated Therapy and Prevention of Thrombosis, 9th ed: American
partial thromboplastin time (APTT) and abnormally low College of Chest Physicians Evidence-Based Clinical Prac-
intrinsic factor (IF) levels. Pathology (Phila) 2015;47:S90. tice Guidelines. Chest 2012;141(2 suppl):e495S-e530S.

January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS Lockshin MD, Kim M, Laskin CA, et al. Prediction of adverse
focused update of the 2014 AHA/ACC/HRS guideline for pregnancy outcome by the presence of lupus anticoagu-
the management of patients with atrial fibrillation: a report lant, but not anticardiolipin antibody, in patients with anti-
of the American College of Cardiology/American Heart phospholipid antibodies. Arthritis Rheum 2012;64:2311-8.
Association Task Force on Clinical Practice Guidelines and Love PE, Santoro SA. Antiphospholipid antibodies: anti-
the Heart Rhythm Society in Collaboration with the Soci- cardiolipin and the lupus anticoagulant in systemic
ety of Thoracic Surgeons. Circulation 2019;140:e125-e151. lupus erythematosus (SLE) and in non-SLE disorders.
Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for Prevalence and clinical significance. Ann Intern Med
VTE disease: CHEST guideline and expert panel report. 1990;112:682-98.
Chest 2016;149:315-52. Lutz J, Menke J, Sollinger D, et al. Haemostasis in chronic
Kelkar AH, Rajasekhar A. Inferior vena cava filters: a kidney disease. Nephrol Dial Transplant 2014;29:29-40.
framework for evidence-based use. Hematology Am Soc Malec K, Broniatowska E, Undas A. Direct oral anticoagu-
Hematol Educ Program 2020;2020:619-28. lants in patients with antiphospholipid syndrome: a cohort
Kelly DM, Ademi Z, Doehner W, et al. Chronic kidney dis- study. Lupus 2020;29:37-44.
ease and cerebrovascular disease: consensus and guid- Martin K, Beyer-Westendorf J, Davidson BL, et al. Use of
ance from a KDIGO Controversies Conference. Stroke direct oral anticoagulants in patients with obesity for
2021;52:e328-e346. treatment and prevention of venous thromboembolism:
Khamashta MA, Cuadrado MJ, Mujic F, et al. The manage- updated communication from the ISTH SSC Subcommit-
ment of thrombosis in the antiphospholipid-antibody tee on Control of Anticoagulation. J Thromb Haemost
syndrome. N Engl J Med 1995;332:993-7. 2021;19:1874-82.

PSAP 2022 Book 1 • Cardiology 198 Anticoagulation in Special Populations


Mavrakanas TA, Garlo K, Charytan DM. Apixaban versus no end-stage renal disease subjects undergoing haemodialy-
anticoagulation in patients undergoing long-term dialy- sis. Thromb Haemost 2015;113:719-27.
sis with incident atrial fibrillation. Clin J Am Soc Nephrol
2020;15:1146-54. Parasrampuria DA, Truitt KE. Pharmacokinetics and pharma-
codynamics of edoxaban, a non-vitamin K antagonist oral
Mavrakanas TA, Samer CF, Nessim SJ, et al. Apixaban phar- anticoagulant that inhibits clotting factor Xa. Clin Pharma-
macokinetics at steady state in hemodialysis patients. cokinet 2016;55:641-55.
J Am Soc Nephrol 2017;28:2241-8.
Pastori D, Menichelli D, Cammisotto V, et al. Use of direct
McAlister FA, Wiebe N, Jun M, et al. Are existing risk scores oral anticoagulants in patients with antiphospho-
for nonvalvular atrial fibrillation useful for prediction or lipid syndrome: a systematic review and comparison
risk adjustment in patients with chronic kidney disease? of the international guidelines. Front Cardiovasc Med
Can J Cardiol 2017;33:243-52. 2021;8:715878.

McDonnell T, Wincup C, Buchholz I, et al. The role of Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus
beta-2-glycoprotein I in health and disease associating warfarin in nonvalvular atrial fibrillation. N Engl J Med
structure with function: more than just APS. Blood Rev 2011;365:883-91.
2020;39:100610.
Pavord S, Myers B. Bleeding and thrombotic complications
McGlasson DL, Romick BG, Rubal BJ. Comparison of a chro- of kidney disease. Blood Rev 2011;25:271-8.
mogenic factor X assay with international normalized ratio
for monitoring oral anticoagulation therapy. Blood Coagul Pengo V, Ruffatti A, Legnani C, et al. Clinical course of high-
Fibrinolysis 2008;19:513-7. risk patients diagnosed with antiphospholipid syndrome.
J Thromb Haemost 2010;8:237-42.
Minet V, Dogné JM, Mullier F. Functional assays in the diag-
nosis of heparin-induced thrombocytopenia: a review. Pengo V, Ruffatti A, Legnani C, et al. Incidence of a first
Molecules 2017;22:617. thromboembolic event in asymptomatic carriers of high-
risk antiphospholipid antibody profile: a multicenter pro-
Miyakis S, Lockshin MD, Atsumi T, et al. International spective study. Blood 2011;118:4714-8.
consensus statement on an update of the classification
criteria for definite antiphospholipid syndrome (APS). Pokorney S. RENal hemodialysis patients ALlocated apix-
J Thromb Haemost 2006;4:295-306. aban versus warfarin in Atrial Fibrillation. November 2019.

Molnar AO, Bota SE, McArthur E, et al. Risk and complica- Proietti M, Boriani G. Obesity paradox in atrial fibrilla-
tions of venous thromboembolism in dialysis patients. tion: implications for outcomes and relationship with
Nephrol Dial Transplant 2018;33:874-80. oral anticoagulant drugs. Am J Cardiovasc Drugs
2020;20:125-37.
Molshatzki N, Orion D, Tsabari R, et al. Chronic kidney
disease in patients with acute intracerebral hemorrhage: Proietti M, Guiducci E, Cheli P, et al. Is there an obesity para-
association with large hematoma volume and poor dox for outcomes in atrial fibrillation? A systematic review
outcome. Cerebrovasc Dis 2011;31:271-7. and meta-analysis of non-vitamin K antagonist oral anti-
coagulant trials. Stroke 2017;48:857-66.
Nagler M, Bakchoul T. Clinical and laboratory tests for the
diagnosis of heparin-induced thrombocytopenia. Thromb Randhawa MS, Vishwanath R, Rai MP, et al. Association
Haemost 2016;116:823-34. between use of warfarin for atrial fibrillation and out-
comes among patients with end-stage renal disease: a
Nilius H, Kaufmann J, Cuker A, et al. Comparative systematic review and meta-analysis. JAMA Netw Open
effectiveness and safety of anticoagulants for the 2020;3:e202175.
treatment of heparin-induced thrombocytopenia.
Am J Hematol 2021;96:805-15. Raskob GE, van Es N, Verhamme P, et al. Edoxaban for the
treatment of cancer-associated venous thromboembo-
Ortel T. Antiphospholipid syndrome laboratory testing and lism. N Engl J Med 2018;378:615-24.
diagnostic strategies. Am J Hematol 2012;87(suppl 1):
S75-S81. Reddel SW, Krilis SA. Testing for and clinical significance
of anticardiolipin antibodies. Clin Diagn Lab Immunol
Ortel TL, Neumann I, Ageno W, et al. American Society of 1999;6:775-82.
Hematology 2020 guidelines for management of venous
thromboembolism: treatment of deep vein thrombosis and Reed D, Palkimas S, Hockman R, et al. Safety and effec-
pulmonary embolism. Blood Adv 2020;4:4693-738. tiveness of apixaban compared to warfarin in dialysis
patients. Res Pract Thromb Haemost 2018;2:291-8.
Ovbiagele B, Wing JJ, Menon RS, et al. Association of
chronic kidney disease with cerebral microbleeds in Reilly PA, Lehr T, Haertter S, et al. The effect of dabigatran
patients with primary intracerebral hemorrhage. Stroke plasma concentrations and patient characteristics on the
2013;44:2409-13. frequency of ischemic stroke and major bleeding in atrial
fibrillation patients: the RE-LY trial (Randomized Evalu-
Parasrampuria DA, Marbury T, Matsushima N, et al. Phar- ation of Long-Term Anticoagulation Therapy). J Am Coll
macokinetics, safety, and tolerability of edoxaban in Cardiol 2014;63:321-8.

PSAP 2022 Book 1 • Cardiology 199 Anticoagulation in Special Populations


Rosove MH, Brewer PM. Antiphospholipid thrombosis: clin- with atrial fibrillation in the United States. Circulation
ical course after the first thrombotic event in 70 patients. 2018;138:1519-29.
Ann Intern Med 1992;117:303-8.
Song S, Kang D, Halim A, et al. Population pharmacokinet-
Ruiz-Irastorza G, Hunt BJ, Khamashta MA. A systematic ic-pharmacodynamic modeling analysis of intrinsic FXa
review of secondary thromboprophylaxis in patients and bleeding from edoxaban treatment. J Clin Pharmacol
with antiphospholipid antibodies. Arthritis Rheum 2014;54:910-6.
2007;57:1487-95.
Sood MM, Bota SE, McArthur E, et al. The three-year
Rumbaut R, Thiagarajan P. Platelet-Vessel Wall Interactions incidence of major hemorrhage among older adults
in Hemostasis and Thrombosis. Morgan & Claypool Life initiating chronic dialysis. Can J Kidney Health Dis
Sciences, 2010. 2014;1:21.

Sammaritano LR. Antiphospholipid syndrome. Best Pract Spyropoulos AC, Brohi K, Caprini J, et al. Scientific and
Res Clin Rheumatol 2020;34:101463. Standardization Committee Communication: guidance
document on the periprocedural management of patients
Sandhu RK, Ezekowitz J, Andersson U, et al. The “obe- on chronic oral anticoagulant therapy: recommendations
sity paradox” in atrial fibrillation: observations from the for standardized reporting of procedural/surgical bleed
ARISTOTLE (Apixaban for Reduction in Stroke and Other risk and patient-specific thromboembolic risk. J Thromb
Thromboembolic Events in Atrial Fibrillation) trial. Eur Haemost 2019;17:1966-72.
Heart J 2016;37:2869-78.
Stangier J, Clemens A. Pharmacology, pharmacokinet-
Saran R, Robinson B, Abbott KC, et al. US Renal Data System ics, and pharmacodynamics of dabigatran etexilate,
2018 annual data report: epidemiology of kidney disease an oral direct thrombin inhibitor. Clin Appl Thromb
in the United States. Am J Kidney Dis 2019;73:A7-A8. 2009;15(1_suppl):9S-16S.
Sarnak MJ, Amann K, Bangalore S, et al. Chronic kidney Stangier J, Rathgen K, Stähle H, et al. Influence of renal
disease and coronary artery disease. J Am Coll Cardiol impairment on the pharmacokinetics and pharmacody-
2019;74:1823-38. namics of oral dabigatran etexilate. Clin Pharmacokinet
Sarratt SC, Nesbit R, Moye R. Safety outcomes of apixaban 2010;49:259-68.
compared with warfarin in patients with end-stage renal Stanton BE, Barasch NS, Tellor KB. Comparison of the
disease. Ann Pharmacother 2017;51:445-50. safety and effectiveness of apixaban versus warfarin in
Schafer JH, Casey AL, Dupre KA, et al. Safety and effi- patients with severe renal impairment. Pharmacotherapy
cacy of apixaban versus warfarin in patients with 2017;37:412-9.
advanced chronic kidney disease. Ann Pharmacother Tektonidou MG, Andreoli L, Limper M, et al. EULAR
2018;52:1078-84. recommendations for the management of anti-
Schenkeveld L, Magro M, Oemrawsingh RM, et al. The phospholipid syndrome in adults. Ann Rheum Dis
influence of optimal medical treatment on the “obesity 2019;78:1296-304.
paradox,” body mass index and long-term mortality in Tittl L, Endig S, Marten S, et al. Impact of BMI on clinical
patients treated with percutaneous coronary intervention: outcomes of NOAC therapy in daily care – results of the
a prospective cohort study. BMJ Open 2012;2:e000535. prospective Dresden NOAC Registry (NCT01588119).
Schulman S, Kakkar AK, Goldhaber SZ, et al. Treatment of Int J Cardiol 2018;262:85-91.
acute venous thromboembolism with dabigatran or warfa- Tripodi A, Chantarangkul V, Clerici M, et al. Laboratory con-
rin and pooled analysis. Circulation 2014;129:764-72. trol of oral anticoagulant treatment by the INR system in
Schulman S, Svenungsson E, Granqvist S. Anticardiolipin patients with the antiphospholipid syndrome and lupus
antibodies predict early recurrence of thromboembolism anticoagulant. Results of a collaborative study involv-
and death among patients with venous thromboembolism ing nine commercial thromboplastins. Br J Haematol
following anticoagulant therapy. Duration of Anticoagula- 2001;115:672-8.
tion Study Group. Am J Med 1998;104:332-8. Upreti VV, Wang J, Barrett YC, et al. Effect of
Schunemann H, Cushman M, Burnett AE, et al. American extremes of body weight on the pharmacokinetics,
Society of Hematology 2018 guidelines for management pharmacodynamics, safety and tolerability of apixaban in
of venous thromboembolism: prophylaxis for hospitalized healthy subjects. Br J Clin Pharmacol 2013;76:908-16.
and nonhospitalized medical patients. Blood Advances van Rein N, Heide-Jørgensen U, Lijfering WM, et al. Major
2018; 2 (22): 3198-3225. bleeding rates in atrial fibrillation patients on single,
Sebaaly J, Kelley D. Direct oral anticoagulants in obesity: dual, or triple antithrombotic therapy. Circulation
an updated literature review. Ann Pharmacother 2019;139:775-86.
2020;54:1144-58. Wang HH, Hung SY, Sung JM, et al. Risk of stroke in long-
Siontis KC, Zhang X, Eckard A, et al. Outcomes associated term dialysis patients compared with the general
with apixaban use in end-stage kidney disease patients population. Am J Kidney Dis 2014;63:604-11.

PSAP 2022 Book 1 • Cardiology 200 Anticoagulation in Special Populations


Wang X, Tirucherai G, Marbury TC, et al. Pharmacokinetics, Washam JB, Holmes DN, Thomas LE, et al. Pharmacotherapy
pharmacodynamics, and safety of apixaban in subjects for atrial fibrillation in patients with chronic kidney
with end-stage renal disease on hemodialysis. J Clin disease: insights from ORBIT-AF. J Am Heart Assoc
Pharmacol 2016;56:628-36. 2018;7:e008928.

Warkentin TE. Laboratory diagnosis of heparin-induced Wattanakit K, Cushman M. Chronic kidney disease
thrombocytopenia. Int J Lab Hematol 2019;41(suppl 1): and venous thromboembolism: epidemiology and
15-25. mechanisms. Curr Opin Pulm Med 2009;15:408-12.

Warkentin TE, Anderson JAM. How I treat patients with a Winkelmayer WC, Patrick AR, Liu J, et al. The increasing
history of heparin-induced thrombocytopenia. Blood prevalence of atrial fibrillation among hemodialysis
2016;128:348-59. patients. J Am Soc Nephrol 2011;22:349-57.

Warkentin TE, Sheppard JI, Moore JC, et al. Quantitative World Health Organization (WHO). Obesity: preventing
interpretation of optical density measurements using and managing the global epidemic. Report of a WHO
PF4-dependent enzyme-immunoassays. J Thromb consultation. World Health Organ Tech Rep Ser 2000;
Haemost 2008;6:1304-12. 894:i-xii, 1-253.

PSAP 2022 Book 1 • Cardiology 201 Anticoagulation in Special Populations


Self-Assessment Questions
13. You are evaluating a patient with a recent deep venous 16. A 52-year-old woman with secondary APS, systemic
thrombosis (DVT) for a potential diagnosis of anti- lupus erythematosus (SLE), and end-stage renal dis-
phospholipid syndrome (APS) who has elevated anti- ease (ESRD) receiving dialysis takes warfarin for second-
cardiolipin antibody (aCL) levels (IgG) greater than 40 ary thromboprophylaxis (target INR 3.0, goal 2.5–3.5).
GPL/MPL units. Which one of the following is best to rec- During dialysis, she develops symptoms consistent with
ommend regarding this patient’s antiphospholipid anti- a transient ischemic attack and is transferred to the ED.
body (APLA) laboratory assessment? Her INR is 2.5. Which one of the following best evaluates
the effectiveness of this patient’s AC therapy?
A. Abnormal APLA laboratory assessments should
be confirmed in 12–14 days to complete the APS A. If the chromogenic factor X (CFX) is 99%, she has
diagnosis. adequate AC to current goal.
B. Abnormal APLA laboratory assessments should B. If the CFX is 33%, she has adequate AC to current
be confirmed in 12 weeks to complete the APS goal.
diagnosis. C. If the activated PTT (aPTT) is 1.5 times the
C. Abnormal APLA antibody assessments do not need laboratory normal, her AC is adequate.
to be reconfirmed to complete the APS diagnosis. D. If the LA is positive, her AC is inadequate because
D. After abnormal APLA laboratory assessments, a the LA interferes with INR accuracy.
dilute Russell viper venom time is recommended
within 2 weeks to complete the APS diagnosis. Questions 17–19 pertain to the following case.

14. Which one of the following patients with APS at low D.A. is a 62-year-old woman in the medical ICU. On rounds, it
bleeding risk would most likely benefit from warfarin is noted that her Plt values have decreased from 110,000/mm3
therapy with a target INR of 3.5 (goal 3.0–4.0)? to 87,000/mm3 overnight. The team suspects heparin-induced
thrombocytopenia (HIT) and wants to order laboratory
A. 25-year-old man with a first provoked venous
testing. You remind D.A.’s care team that it would be best to
thrombotic event (venous thromboembolism [VTE])
use the 4T score to determine the need for HIT testing.
with obesity who is positive for lupus anticoagulant
(LA) 17. Which one of the following best justifies use of the 4T
B. 32-year-old woman with one episode of obstetric score for D.A.?
APS 18 months ago being seen today for A. It has a high positive predictive value and is good for
preconception counseling ruling in HIT.
C. 35-year-old man with a high-risk APLA profile but no B. It is the most validated pretest clinical probability
personal history of thrombosis (PTCP) score for HIT.
D. 42-year-old woman with secondary APS presenting C. It ranges from 0 to 8 points, depending on highly
with a recurrent DVT on warfarin at an INR of 2.6 objective criteria.
15. The primary care provider for a 45-year-old man with a D. A score of 3 is considered moderate risk and
new episode of VTE requests an anticoagulation (AC) warrants enzyme-linked immunosorbent assay
consult because there is high suspicion for APS. Which (ELISA) testing.
one of the following best evaluates the current evidence 18. D.A.’s care team calculates her 4T score as 4, and testing
for use of rivaroxaban in treating and preventing throm- is recommended. Which one of the following is best to
boembolism if this patient is confirmed to have APS? recommend regarding laboratory testing for HIT type 2
A. Has high-quality safety and efficacy data in patients in D.A.?
with low-risk APS, and data are promising in high- A. Serotonin release assay (SRA) is a highly specific
risk APS. functional assay and is considered the gold
B. Has limited safety and efficacy in both low- and high- standard.
risk APS, with current guidance to avoid use in APS. B. Heparin-induced platelet aggregation assay (HIPA)
C. Was shown noninferior to warfarin in patients is an immunoassay that detects platelet factor 4
with triple-positive thrombotic APS as a safe and (PF4)/heparin antibodies.
effective therapeutic option. C. ELISA is a highly specific functional assay that is
D. Was shown superior to warfarin in patients with reported in optical density (OD).
triple-positive thrombotic APS as a safe and D. SRA has a rapid turnaround time and should be used
effective therapeutic option. first line.

PSAP 2022 Book 1 • Cardiology 202 Anticoagulation in Special Populations


19. D.A. is stable but requires AC therapy while the team catheter-directed thrombolysis and is transferred from
awaits laboratory results. On the basis of the ASH 2018 the cardiac ICU to the progressive care ward on hospi-
HIT guidelines, which one of the following is best to rec- tal day 4. Your antithrombosis stewardship service is
ommend for D.A.? consulted for recommendations on oral anticoagulant
(OAC) therapies for this patient. On interview, the patient
A. Argatroban is preferred for initial therapy in all
describes having what sounds like an unprovoked prox-
patients with HIT because it is FDA approved.
imal DVT last year. He was prescribed warfarin at that
B. Direct oral anticoagulants (DOACs) are preferred to
time; however, he took it for only a few weeks because
warfarin, but regimens used for isolated HIT differ
he lives more than 130 miles from the nearest medical
from those used in clinic trials.
facility and could not make frequent trips to get his INR
C. Patients with suspected HIT should immediately be
checked. He sees a primary care provider at a rural clinic
initiated on prophylactic-intensity non-heparin AC
two or three times per year who prescribes his medica-
therapy.
tions, including glipizide, furosemide, losartan, and ator-
D. All patients with HIT should receive 3–6 months of
vastatin. He is otherwise ready for discharge and eager
AC therapy.
to go home. Which one of the following is best to recom-
20. A 78-year-old woman (height 66 inches, weight 61 kg) mend for this patient’s recurrent unprovoked VTE?
presents to the urgent care clinic for shortness of breath
A. Place a retrievable inferior vena cava (IVC) filter and
and a feeling of “butterflies” in her chest that began this
have him follow up with his primary care provider in
morning while doing laundry. A 12-lead ECG reveals atrial
3 months for evaluation for removal.
flutter with heart rate 102 beats/minute. The patient’s
B. Discontinue heparin and prescribe apixaban 5 mg
medical history includes hypertension, diabetes, glau-
orally twice daily for a total duration of 3 months
coma, stage 5 CKD, and peripheral arterial disease
with monthly follow-up.
requiring a femoral-popliteal bypass graft in 2017. She
C. Discontinue heparin and prescribe apixaban 10
also reports occasional hemorrhoid-associated bleed-
mg orally twice daily for 3 more days, then 5 mg
ing. She currently takes aspirin 81 mg daily, lisinopril
twice daily for at least 3 months followed by regular
5 mg daily, atorvastatin 20 mg, basal and bolus insulin,
assessment for tolerance.
and latanoprost; her SCr is 1.9 mg/dL. Through shared
D. Keep him in the hospital for continued
decision-making, she expresses significant concern
heparinization as a bridge to warfarin until his
about having a stroke and would prefer preventive ther-
INR is greater than 2 and tell him to arrange self-
apy. Which one of the following is best to recommend for
management of INR at home.
this patient?
22. As the AC stewardship pharmacist for your hospital, you
A. Continue aspirin for both stroke prevention and
have been asked to provide medical grand rounds on the
peripheral arterial disease.
current use of DOACs in the treatment of atrial fibrillation
B. Initiate apixaban 2.5 mg twice daily and discuss
(AF) and VTE events. Which one of the following best
aspirin with her vascular surgeon.
describes the current data for DOAC use in patients at
C. Initiate apixaban 5 mg twice daily and discuss
extremes of weight?
aspirin with her vascular surgeon.
D. Initiate warfarin and continue aspirin. A. Large, well-controlled randomized clinical trials
(RCTs) have been conducted to evaluate DOAC
21. A 48-year-old man (height 67 inches, weight 88 kg, BMI
safety and efficacy.
30.4 kg/m2) with ESRD not yet on dialysis as well as dia-
B. The number of underweight patients and patients
betes and hypertension is transferred from an outside
with obesity included in phase III clinical trials have
hospital to your ED by MedFlight for bilateral submas-
been limited to less than 10% and 40%, respectively.
sive pulmonary embolisms (PEs). He was initiated on a
C. The definitions of obesity were homogeneous in the
heparin infusion at the outside hospital, but no throm-
currently available clinical trial data.
bolytics were administered. Laboratory tests confirm an
D. All trials included formal, standardized safety
elevated BNP of 2260 pg/mL and increased troponins.
and efficacy evaluations as a primary end point in
A CT chest and cardiac echocardiogram reveal a right
patients at weight extremes.
ventricular/left ventricular ratio of 1.8, and his esti-
mated CrCl is 13 mL/minute. He successfully undergoes

PSAP 2022 Book 1 • Cardiology 203 Anticoagulation in Special Populations


23. A 63-year-old man (weight 182 kg, BMI 56 kg/m2) devel- 24. A 66-year-old man (height 69 inches, weight 55 kg, BMI
ops proximal DVT following emergency orthopedic sur- 19.5 kg/m2) presents to his primary care provider with
gery after an acute ankle fracture with surgical repair. He chief concerns of shortness of breath and tachycardia
has no history of thrombotic events. His baseline creat- and is subsequently diagnosed with AF. The patient’s
inine and hematologic profile are normal. The patient is medical history includes hypertension and mild hyperlip-
adamantly against the use of warfarin. Which one of the idemia, both being treated appropriately. His renal func-
following is the best rivaroxaban dosing to recommend tion is normal (CrCl 70 mL/minute), and he is at average
fo this patient? bleeding risk. Which one of the following is best to rec-
ommend for this patient’s initial AC therapy?
A. Rivaroxaban 10 mg orally once daily
B. Rivaroxaban 15 mg orally once daily A. Apixaban 2.5 mg orally twice daily
C. 15 mg orally twice daily for 3 weeks, then lower to 10 B. Aspirin 650 mg orally twice daily
orally once daily C. Edoxaban 30 mg orally once daily
D. 15 mg orally twice daily for 3 weeks, then lower to 20 D. Warfarin 5 mg orally daily (goal INR 2.0–3.0)
orally once daily

PSAP 2022 Book 1 • Cardiology 204 Anticoagulation in Special Populations


Learner Feature Evaluation: Interactive Case: Anticoagulation in Special
Populations

As you take the posttest for this feature, also evaluate the 22. The content of the feature was useful to me.
material’s quality and usefulness, as well as the achieve- 23. The teaching and learning methods used in the feature
ment of learning objectives. Rate each item using this 5-point were effective.
scale: 24. The active learning methods used in the feature were
effective.
• Strongly agree
• Agree 25. The learning assessment activities used in the feature
• Neutral were effective.
• Disagree 26. The feature was effective overall.
• Strongly disagree 27. The activity met the stated learning objectives.
16. The content of the feature met my educational needs. 28. If any objectives were not met, please list them here.
17. The content of the feature satisfied my expectations.
18. The author presented the feature content effectively. OTHER COMMENTS
19. The content of the feature was relevant to my practice 29. Please provide any specific comments related to any
and presented at the appropriate depth and scope. perceptions of bias, promotion, or advertisement of
20. The content of the feature was objective and balanced. commercial products.

21. The content of the feature is free of bias, promotion, and 30. Please expand on any of your above responses, and/or
advertisement of commercial products. provide any additional comments regarding this feature:

PSAP 2022 Book 1 • Cardiology 205 Anticoagulation in Special Populations


Interactive Case: Cardiovascular
Diseases in Pregnancy
By Lindsey Federle, Pharm.D., BCCP, BCPS

Reviewed by Jessie Dunne, Pharm.D., BCPS, BCCP; and Leslie Wooten, Pharm.D., BCPS

LEARNING OBJECTIVES

1. Evaluate patient risk factors for cardiovascular complications during pregnancy and stratify according to the modified
WHO pregnancy risk classes.
2. Distinguish the physiologic and pharmacokinetic changes that occur in pregnancy.
3. Assess hypertensive disorders of pregnancy and devise treatment plans throughout pregnancy.
4. Develop an appropriate medical regimen for peripartum cardiomyopathy.
5. Design safe and effective anticoagulant regimens during pregnancy.
6. Develop pharmacotherapy to manage acute arrhythmia in the pregnant patient.

ABBREVIATIONS IN THIS FEATURE


ACOG American College of Obstetricians BASELINE KNOWLEDGE STATEMENTS
and Gynecologists
Readers of this feature are presumed to be familiar with the
CVD Cardiovascular disease
following:
HDP Hypertensive disorder of
pregnancy • Terminology in pregnancy
LVEF Left ventricular ejection fraction • Basic pharmacokinetic and pharmacodynamic principles,
including absorption, bioavailability, metabolism and
PPCM Peripartum cardiomyopathy transport, and excretion
SMM Severe maternal morbidity • General knowledge of cardiovascular medication dosing,
SVT Supraventricular tachycardia adverse effects, and pharmacokinetics
• Management of hypertension, heart failure, venous throm-
Table of other common abbreviations. boembolism, valvular heart disease, and arrhythmias in a
non-pregnant population
Table of common laboratory reference values

ADDITIONAL READINGS

The following resources have additional background informa-


tion on this topic:
• Halpern DG, Weinberg CR, Pinnelas R, et al. Use of medica-
tion for cardiovascular disease during pregnancy: JACC
state-of-the-art review. J Am Coll Cardiol 2019;73:457-76.
• Mehta LS, Warnes CA, Bradley E, et al. Cardiovascular con-
siderations in caring for pregnant patients: a scientific
statement from the American Heart Association. Circula-
tion 2020;141:e884-e903.
• Regitz-Zagrosek V, Roos-Hesselink JW, Bauersachs J, et al.
2018 ESC guidelines for the management of cardiovascular
diseases during pregnancy. Eur Heart J 2018;39:3165-241.
• Online Appendix

PSAP 2022 Book 1 • Cardiology 207 Interactive Case: Cardiovascular Diseases in Pregnancy
American College of Obstetricians and Gynecologists (ACOG)
Practice Points Practice Bulletin: pregnancy and heart disease. Obstet
• Pregnancy causes significant physiologic and pharmacoki- Gynecol 2019b;133:e320-56.
netic changes. Medication management of cardiovascular
American College of Obstetricians and Gynecologists (ACOG)
disease (CVD) in pregnancy presents many challenges.
Practice Bulletin: gestational hypertension and
• Women who have underlying CVD or who develop cardio-
preeclampsia. Obstet Gynecol 2020;135:e237-60.
vascular conditions are at an increased risk of morbidity
and mortality and should be assessed before pregnancy for Bayer-Westendorf J, Marten S. Reproductive issues in women
counseling, medication management, and stabilization of on direct oral anticoagulants. Res Pract Thromb Haemost
underlying disease state. 2021;5:e12512.
• Hypertensive disorders of pregnancy (HDPs) are a hetero-
geneous group of disorders that can worsen at any time. Caruso G, Scopelliti A, Scaramuzzino S, et al. Cabergoline
No consensus exists regarding blood pressure goals in as an adjuvant to standard heart failure treatment in peri-
managing HDPs. partum cardiomyopathy: a case report and review of the
• If preeclampsia develops, gestational age and presence of literature. Case Rep Womens Health 2021;29:e00277.
severe features will guide management decisions regarding
Casele HL, Laifer SA, Woelkers DA, et al. Changes in the
delivery of care and initiation of magnesium sulfate.
pharmacokinetics of the low-molecular-weight hepa-
• Until the pathophysiology of peripartum cardiomyopathy
rin enoxaparin sodium during pregnancy. Am J Obstet
(PPCM) is better understood, medical management will
Gynecol 1999;181(5 pt 1):1113-7.
overlap with that for other types of heart failure to achieve
goals of improving left ventricular function and preventing Centers for Disease Control and Prevention (CDC). Preg-
morbidity and mortality. Unique considerations for PPCM nancy Mortality Surveillance System. 2020. Available
include stage of pregnancy (i.e., antepartum or postpar- at https://www.cdc.gov/reproductivehealth/maternal-­
tum), whether there is a desire to breastfeed, and whether mortality/pregnancy-mortality-surveillance-system.htm.
bromocriptine or anticoagulation should be prescribed.
• Anticoagulation in pregnancy presents a challenge in bal- Centers for Disease Control and Prevention (CDC) Foundation.
ancing therapeutic efficacy with antithrombotic safety for Building U.S. Capacity to Review and Prevent Maternal
both the patient and the developing fetus. When possible, Deaths. 2018. Report from Nine Maternal Mortality Review
anticoagulation planning should begin before conception Committees. Available at https://www.cdcfoundation.org/
to allow for patient and provider discussion and documen- sites/default/files/files/ReportfromNineMMRCs.pdf.
tation of plan, particularly for patients with mechanical Drenthen W, Boersma E, Balci A, et al. Predictors of pregnancy
heart valves.
complications in women with congenital heart disease.
• New-onset arrhythmia in pregnancy should prompt an
ZAHARA Investigators. Eur Heart J 2010;31:2124-32.
evaluation for underlying conditions. Antiarrhythmic agents
should be avoided, if possible; however, when used, the D’Souza R, Ostro J, Shah PS, et al. Anticoagulation for preg-
lowest recommended dose should be initiated, with ad- nant women with mechanical heart valves: a systematic
justments made according to response. If maternal cardiac review and meta-analysis. Eur Heart J 2017;38:1509-16.
arrest occurs, causes of arrest should be considered and
the clinician should not hesitate to administer medications,
Duley L, Gulmezoglu AM, Henderson-Smart DJ, et al. Mag-
given the gravity of the situation. nesium sulphate and other anticonvulsants for women
with pre-eclampsia. Cochrane Database Syst Rev
2010;11:CD000025.
Duley L, Meher S, Hunter KE, et al. Antiplatelet agents for
INTERACTIVE CASE: preventing preeclampsia and its complications. Cochrane
CARDIOVASCULAR DISEASES IN Database Syst Rev 2019;10:CD004659.
PREGNANCY
Hilfiker-Kleiner D, Haghikia A, Berliner D, et al. Bromocriptine
• Click here to start this PSAP activity. for the treatment of peripartum cardiomyopathy: a multi-
centre randomized study. Eur Heart J 2017;38:2671-9.
Hoffman MK, Goudar SS, Kodkany BS, et al.; ASPIRIN Study
REFERENCES Group. Low-dose aspirin for the prevention of preterm
Altman D, Carroli G, Duley L, et al. Do women with preeclamp- delivery in nulliparous women with a singleton pregnancy
sia, and their babies, benefit from magnesium sulphate? (ASPIRIN): a randomised, double-blind, placebo-controlled
The Magpie trial: a randomised placebo-controlled trial. trial. Lancet 2020;395:285-93.
Lancet 2002;359:1877-90.
Hofmeyr GJ, Lawrie TA, Atallah AN, et al. Calcium supple-
American College of Obstetricians and Gynecologists mentation during pregnancy for preventing hypertensive
(ACOG) Practice Bulletin: thromboembolism in pregnancy. disorders and related problems. Cochrane Database Syst
Obstet Gynecol 2018;132:e1-e17. Rev 2018;10:CD001059.
American College of Obstetricians and Gynecologists (ACOG) Jeejeebhoy FM, Zelop CM, Lipman S, et al. Cardiac arrest
Practice Bulletin: chronic hypertension in pregnancy. in pregnancy: a scientific statement from the American
Obstet Gynecol 2019a;133:e26-e50. Heart Association. Circulation 2015;132:1747-73.

PSAP 2022 Book 1 • Cardiology 208 Interactive Case: Cardiovascular Diseases in Pregnancy
Koczo A, Marino A, Jeyabalan A. Breastfeeding, cellular Rosenbloom JI, Sabol BA, Chung C, et al. Improving medi-
immune activation, and myocardial recovery in peripartum cation error identification with an inpatient maternal-fetal
cardiomyopathy. JACC Basic Transl Sci 2019;4:291-300. medicine pharmacist. Poster presented at: 39th Annual
Pregnancy Meeting, Society for Maternal-Fetal Medicine;
Magee LA, von Dadelszen P, Rey E, et al. Less-tight versus February 2019; Las Vegas, NV.
tight control of hypertension in pregnancy. N Engl J Med
2015;372:407-17. Scaffidi J, Mol BW, Keelan JA. The pregnant women as a
drug orphan: a global survey of registered clinical trials
Magee LA, von Dadelszen P, Singer J, et al. The CHIPS ran- of pharmacological interventions in pregnancy. BJOG
domized controlled trial (Control of Hypertension in 2017;124:132-40.
Pregnancy Study): is severe hypertension just an elevated
blood pressure? Hypertension 2016;68:1153-9. Silversides CK, Grewal J, Mason J, et al. Pregnancy out-
comes in women with heart disease: the CARPREG II
Malhame I, Danilack VA, Raker CA, et al. Cardiovascu- study. J Am Coll Cardiol 2018;71:2419-30.
lar severe maternal morbidity in pregnant and postpar-
tum women: development and internal validation of risk Sliwa K, Blauwet L, Tibazarwa K, et al. Evaluation of bro-
prediction models. BJOG 2021;128:922-32. mocriptine in the treatment of acute severe peripartum
cardiomyopathy: a proof-of-concept pilot study. Circula-
Otto CM, Nishimura RA, Bonow RO, et al. 2020 ACC/AHA tion 2010;121:1465-73.
guideline for the management of patients with valvular
heart disease. Circulation 2021;143:e72-e227. Stephenson ML, Serra AE, Neeper JM. A randomized con-
trolled trial of differing doses of post-cesarean enoxa-
Ozkan M, Cakal B, Karakoyun S, et al. Thrombolytic therapy parin thromboprophylaxis in obese women. J Perinatol
for the treatment of prosthetic heart valve thrombosis 2016;36:95-9.
in pregnancy with low-dose, slow infusion of tissue-type
plasminogen activator. Circulation 2013;128:532-40. Thorne S, MacGregor A, Nelson-Piercy C. Risks of
contraception and pregnancy in heart disease. Heart
Parikh NI, Gonzalez JM, Anderson CAM, et al. Adverse preg- 2006;92:1520-5.
nancy outcomes and cardiovascular disease risk: unique
opportunities for cardiovascular disease prevention in von Dadelszen P, Payne B, Li J, et al. Prediction of adverse
women: a scientific statement from the American Heart maternal outcomes in pre-eclampsia: development and
Association. Circulation 2021;143:e903-16. validation of the fullPIERS model. Lancet 2011;377:219-27.

Pfaller B, Sathananthan G, Grewal J, et al. Preventing compli- Yaméogo NV, Kagambèga LJ, Seghda A, et al. Bromocriptine
cations in pregnant women with cardiac disease. in management of peripartum cardiomyopathy: a random-
J Am Coll Cardiol 2020;75:1443-52. ized study on 96 women in Burkina Faso. J Cardiol Clin
Res 2017;5:1098-106.
Pinheiro EA, Stika CS. Drugs in pregnancy: pharmacologic
and physiologic changes that affect clinical care. Semin Yucel E, Yeh DD. Pregnancy in women with congenital heart
Perinatol 2020;44:151221. disease. Curr Treat Options Cardiovasc Med 2017;19:73.

Rashba EJ, Zareba W, Moss AJ, et al. Influence of pregnancy Wolfe DS, Hameed AB, Taub CC, et al. Addressing mater-
on the risk for cardiac events in patients with heredi- nal mortality: the pregnant cardiac patient. Am J Obstet
tary long QT syndrome. LQTS Investigators. Circulation Gynecol 2019;220:167.e1-8.
1998;97:451-6.
Zeisler H, Llurba E, Chantraine F, et al. Predictive value of the
Regitz-Zagrosek V, Roos-Hesselink JW, Bauersachs J, et al. sFlt-1:PlGF ratio in women with suspected preeclampsia.
2018 ESC guidelines for the management of cardio- N Engl J Med 2016;374:130-22.
vascular diseases during pregnancy. Eur Heart J
2018;39:3165-241.

Rolnik DL, Wright D, Poon LC, et al. aspirin versus placebo in


pregnancies at high risk for preterm preeclampsia. N Engl
J Med 2017;377:613-22.

PSAP 2022 Book 1 • Cardiology 209 Interactive Case: Cardiovascular Diseases in Pregnancy
Self-Assessment Questions
Questions 25–27 pertain to the following case. 28. Which one of the following women is at highest risk
R.W., a 28-year-old woman with a medical history of severe of maternal morbidity and mortality when becoming
rheumatic mitral stenosis with valve replacement 2 months pregnant?
ago, is contemplating pregnancy. Her home drugs include
A. 27-year-old with long QT syndrome and history of
metoprolol 25 mg orally twice daily, and warfarin 5 mg
ventricular tachycardia
alternating with 2.5 mg each day. R.W.’s ECHO after sur-
B. 28-year-old with unrepaired severe asymptomatic
gery revealed ejection fraction (EF) 45%, well-functioning
aortic stenosis
mechanical mitral valve, and mild aortic stenosis. Her vital
C. 35-year-old with a history of peripartum
signs are stable, and she is in no apparent distress. R.W.’s
cardiomyopathy (PPCM) (current EF 25%)
laboratory values include INR 2.6 and SCr 0.6 mg/dL.
D. 40-year-old with hypertension undergoing assisted
25. Which one of the following is the best frequency of moni- reproduction
toring by a cardiologist to recommend throughout R.W.’s
29. A 25-year-old woman with congenital long QT syndrome
pregnancy?
is 8 weeks pregnant. Her vital signs include blood pres-
A. Weekly sure 106/62 mm Hg and heart rate 72 beats/minute.
B. Every 2 weeks Which one of the following is best to recommend initiat-
C. Monthly ing in this patient?
D. Once each trimester
A. Metoprolol
26. For which one of the following physiologic alterations is B. Atenolol
R.W. most at risk during her pregnancy? C. Carvedilol
A. Increases in total body volume and cardiac output D. Propranolol
will alter her valve gradients and worsen stenotic
physiology. Questions 30–32 pertain to the following case.

B. Anticoagulation during pregnancy will increase her H.T., a 42-year-old woman (height 68 inches, weight 77 kg),
risk of anemia requiring iron replacement. has a medical history that includes a single pulmonary embo-
C. Physiologic changes of pregnancy will alter her lism (PE), a factor V Leiden homozygote, and hypertension.
warfarin pharmacokinetics and decrease her dose She is nulliparous and 5 weeks pregnant after in vitro fertil-
requirement. ization. H.T.’s home drugs include apixaban 5 mg twice daily
D. Hypercoagulable state of pregnancy will increase and nifedipine 30 mg daily. Her vital signs include blood pres-
her risk of valve-related thrombosis. sure 145/88 mm Hg and heart rate 84 beats/minute. Her labo-
ratory values include BUN 18 mg/dL, SCr 0.8 mg/dL, Hgb 15.2
27. Which one of the following educational points regarding
g/dL, Plt 208,000/mm3, and protein/creatinine ratio 0.2
anticoagulation management during pregnancy is best
mg/dL.
to provide to R.W.?
30. Which one of the following is best to recommend to man-
A. Changing to enoxaparin 1 mg/kg subcutaneously
age H.T.’s anticoagulation?
every 12 hours now will minimize the risk of
teratogenicity. A. Continue apixaban 5 mg twice daily for all
B. Continuing warfarin throughout pregnancy to target trimesters.
an INR of 2.5–3.5 will minimize the risk of valvular B. Administer enoxaparin 40 mg subcutaneously every
thrombosis. 12 hours for all trimesters.
C. Targeting an INR of 1.5–2.5 throughout pregnancy C. Administer enoxaparin 80 mg subcutaneously every
can minimize the risk of teratogenicity and prevent 12 hours for all trimesters.
valvular thrombosis. D. Administer enoxaparin 80 mg subcutaneously
D. Placing a bioprosthetic valve will minimize the every 12 hours for the first trimester, then change to
maternal risk of thrombosis and fetal toxicity from warfarin in the second and third trimesters.
anticoagulation during pregnancy.

PSAP 2022 Book 1 • Cardiology 210 Interactive Case: Cardiovascular Diseases in Pregnancy
31. H.T., now 28 weeks 4 days pregnant, visits the office for a vaginal delivery. Immediately after delivery, a maternal car-
blood pressure check with no other concerns. Her medi- diac arrest was called for ventricular fibrillation. Before the
cations include nifedipine 60 mg daily and labetalol 400 cardiac arrest, J.R. was being treated for preeclampsia with a
mg three times daily. Her vital signs include blood pres- magnesium sulfate continuous infusion 2 g/hour and labeta-
sure 154/92 mm Hg and heart rate 92 beats/minute. Her lol 100 mg twice daily. She is currently 6 minutes into car-
laboratory values include SCr 1.1 mg/dL, Hgb 13.6 g/dL, diopulmonary resuscitation and has received defibrillation
Plt 154,000/mm3, AST 36 U/L, ALT 20 U/L, LDH 186 U/L, twice, epinephrine 1 mg intravenously every 3 minutes, and
and protein/creatinine ratio 0.23 mg/dL. Which one of amiodarone 300 mg intravenously once; magnesium sulfate
the following best assesses H.T.’s hypertensive disorder was discontinued.
of pregnancy (HDP)?
34. Which one of the following is best to recommend to J.R.’s
A. Chronic hypertension maternal cardiac arrest team?
B. Chronic hypertension with superimposed
A. Amiodarone should only be administered after other
preeclampsia
medications have failed.
C. Gestational hypertension
B. Central access must be above the diaphragm for
D. Preeclampsia
medication administration.
32. H.T., now 34 weeks 2 days pregnant, has been monitor- C. Vasopressin 40 units once is preferred to assist with
ing her blood pressure and reports a result of 172/110 uterine contraction.
mm Hg and a similar value repeated after 15 minutes. D. Calcium chloride 1 g intravenously should be
She had a headache this morning, but acetaminophen administered.
resolved her symptoms. Her medications include nifed-
35. J.R. achieves return of spontaneous circulation after 9
ipine 90 mg daily and labetalol 400 mg three times daily.
minutes, and her neurologic function is intact. Workup
Which one of the following is best to recommend for
of her cardiac arrest reveals an EF of 20% and pulmonary
managing H.T.’s blood pressure?
edema on chest radiography, with no coronary artery dis-
A. Methyldopa ease seen on coronary angiogram. Her laboratory values
B. Intravenous hydralazine are SCr 0.6 mg/dL, K 4.6 mEq/L, magnesium 5.6 mEq/L,
C. Intravenous magnesium sulfate and N-terminal pro–brain natriuretic peptide 16,204
D. Immediate-release nifedipine pg/mL (range 15–450 pg/mL). J.R.’s vital signs include
blood pressure 146/84 mm Hg, heart rate 110 beats/min-
33. A 32-year-old nulliparous woman (height 66 inches,
ute, respiratory rate 24 breaths/minute, and Sao2 89%.
weight 86 kg) is 12 weeks 2 days’ gestation. Her medical
Which one of the following, together with furosemide 40
history includes type 1 diabetes, hypertension, and a pro-
mg intravenously once, is best to recommend initiating
thrombin G20210A mutation heterozygote. Her grand-
for suspected PPCM in J.R.?
mother had a PE at age 42; the patient has no history
of venous thromboembolism (VTE). Today’s examination A. Hydralazine 10 mg intravenously once
shows the patient has blood pressure 135/85 mm Hg and B. Nitroglycerin infusion 5–200 mcg/minute
heart rate 80 beats/minute. Her home drugs include con- C. Metoprolol 5 mg intravenously once
tinuous glucose monitoring with an insulin pump and D. Milrinone infusion 0.25 mcg/kg/minute
labetalol 200 mg twice daily. Which one of the following
36. Which one of the following educational points is best to
is best to recommend to improve this patient’s maternal
share with J.R. regarding bromocriptine use for PPCM?
outcomes?
A. Bromocriptine will improve left ventricular function
A. Add aspirin 81 mg daily.
and decrease mortality.
B. Add calcium carbonate 500 mg three times daily.
B. Breastfeeding will not be feasible if prescribed
C. Add enoxaparin 40 mg subcutaneously daily.
bromocriptine.
D. Increase labetalol to 300 mg twice daily.
C. Longer durations of bromocriptine show superior
outcomes in improving EF.
Questions 34–36 pertain to the following case. D. Anticoagulation will not be needed if she does not
J.R. is a 22-year-old woman with no contributory medi- use bromocriptine.
cal history. She was being treated for preeclampsia with
severe features when she gave birth at 36 weeks 2 days by

PSAP 2022 Book 1 • Cardiology 211 Interactive Case: Cardiovascular Diseases in Pregnancy
Learner Feature Evaluation: Interactive Case: Cardiovascular Diseases
in Pregnancy

As you take the posttest for this feature, also evaluate the 4 0. The learning assessment activities used in the feature
material’s quality and usefulness, as well as the achievement were effective.
of learning objectives. Rate each item using this 5-point scale: 41. The feature was effective overall.

• Strongly agree 42. The activity met the stated learning objectives.
• Agree 4 3. If any objectives were not met, please list them here.
• Neutral
• Disagree OTHER COMMENTS
• Strongly disagree
4 4. Please provide any specific comments related to any
31. The content of the feature met my educational needs. perceptions of bias, promotion, or advertisement of
32. The content of the feature satisfied my expectations. commercial products.
33. The author presented the feature content effectively. 45. Please expand on any of your above responses, and/or
provide any additional comments regarding this feature:
34. The content of the feature was relevant to my practice
and presented at the appropriate depth and scope.
Questions 46–48 apply to the entire learning module.
35. The content of the feature was objective and balanced.
46. How long did it take you to read the instructional materi-
36. The content of the feature is free of bias, promotion, and als in this module?
advertisement of commercial products.
47. How long did it take you to read and answer the assess-
37. The content of the feature was useful to me. ment questions in this module?
38. The teaching and learning methods used in the feature 4 8. Please provide any additional comments you may have
were effective. regarding this module:
39. The active learning methods used in the feature were
effective.

PSAP 2022 Book 1 • Cardiology 212 Interactive Case: Cardiovascular Diseases in Pregnancy

You might also like