You are on page 1of 467

Al-Khatib SM, et al.

2017 VA/SCD Guideline: Executive Summary

2017 AHA/ACC/HRS Guideline for Management of Patients With


Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death:
Executive Summary

A Report of the American College of Cardiology/American Heart Association Task Force on


Clinical Practice Guidelines and the Heart Rhythm Society

Developed in Collaboration With the Heart Failure Society of America

WRITING COMMITTEE MEMBERS


Sana M. Al-Khatib, MD, MHS, FACC, FAHA, FHRS, Chair
William G. Stevenson, MD, FACC, FAHA, FHRS, Vice Chair*
Michael J. Ackerman, MD, PhD* Anne M. Gillis, MD, FHRS*
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

William J. Bryant, JD, LLM Mark A. Hlatky, MD, FACC, FAHA


David J. Callans, MD, FACC, FHRS* Christopher B. Granger, MD, FACC, FAHA*
Anne B. Curtis, MD, FACC, FAHA, FHRS* Stephen C. Hammill, MD, FACC, FHRS
Barbara J. Deal, MD, FACC, FAHA Jos A. Joglar, MD, FACC, FAHA, FHRS
Timm Dickfeld, MD, PhD, FHRS* G. Neal Kay, MD
Michael E. Field, MD, FACC, FAHA, FHRS Daniel D. Matlock, MD, MPH
Gregg C. Fonarow, MD, FACC, FAHA, FHFSA* Robert J. Myerburg, MD, FACC
Richard L. Page, MD, FACC, FAHA, FHRS

ACC/AHA TASK FORCE MEMBERS


Glenn N. Levine, MD, FACC, FAHA, Chair
Patrick T. OGara, MD, MACC, FAHA, Chair-Elect
Jonathan L. Halperin, MD, FACC, FAHA, Immediate Past Chair
Sana M. Al-Khatib, MD, MHS, FACC, FAHA Federico Gentile, MD, FACC
Joshua A. Beckman, MD, MS, FAHA Samuel Gidding, MD, FAHA
Kim K. Birtcher, MS, PharmD, AACC Zachary D. Goldberger, MD, MS, FACC, FAHA
Biykem Bozkurt, MD, PhD, FACC, FAHA Mark A. Hlatky, MD, FACC, FAHA
Ralph G. Brindis, MD, MPH, MACC John Ikonomidis, MD, PhD, FAHA
Joaquin E. Cigarroa, MD, FACC Jos A. Joglar, MD, FACC, FAHA
Anita Deswal, MD, MPH, FACC, FAHA Laura Mauri, MD, MSc, FAHA
Lesley H. Curtis, PhD, FAHA Barbara Riegel, PhD, RN, FAHA
Lee A. Fleisher, MD, FACC, FAHA Susan J. Pressler, PhD, RN, FAHA
Duminda N. Wijeysundera, MD, PhD

*Writing committee members are required to recuse themselves from voting on sections to which their specific
relationships with industry may apply; see Appendix 1 for detailed information. Section numbers pertain to those in
the full-text guideline. ACC/AHA Representative. HRS Representative. ACC/AHA Task Force on Performance
Measures Liaison/HFSA Representative. ACC/AHA Task Force on Clinical Practice Guidelines Liaison. Former Task
Force member; current member during the writing effort.

This document was approved by the American College of Cardiology Clinical Policy Approval Committee, the American
Heart Association Science Advisory and Coordinating Committee, and the Heart Rhythm Society in September 2017,
and the American Heart Association Executive Committee in October 2017.

Page 1
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

The Comprehensive RWI Data Supplement table is available with this article at
http://circ.ahajournals.org/lookup/suppl/doi:10.1161/CIR.0000000000000548/-/DC1.

The online Data Supplement is available with this article at


http://circ.ahajournals.org/lookup/suppl/doi:10.1161/CIR.0000000000000548/-/DC2.

The American Heart Association requests that this document be cited as follows: Al-Khatib SM, Stevenson WG,
Ackerman MJ, Bryant WJ, Callans DJ, Curtis AB, Deal BJ, Dickfeld T, Field ME, Fonarow GC, Gillis AM, Hlatky MA, Granger
CB, Hammill SC, Joglar JA, Kay GN, Matlock DD, Myerburg RJ, Page RL. 2017 AHA/ACC/HRS guideline for management of
patients with ventricular arrhythmias and the prevention of sudden cardiac death: executive summary: a report of the
American College of Cardiology Foundation/American Heart Association Task Force on Clinical Practice Guidelines and
the Heart Rhythm Society. Circulation. 2017;:ee. DOI: 10.1161/CIR.0000000000000548.

This article has been copublished in the Journal of the American College of Cardiology and HeartRhythm.

Copies: This document is available on the World Wide Web sites of the American College of Cardiology (www.acc.org),
the American Heart Association (professional.heart.org), and the Heart Rhythm Society (www.hrsonline.org). A copy of
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

the document is available at http://professional.heart.org/statements by using either Search for Guidelines &
Statements or the Browse by Topic area. To purchase additional reprints, call 843-216-2533 or e-mail
kelle.ramsay@wolterskluwer.com.

Expert peer review of AHA Scientific Statements is conducted by the AHA Office of Science Operations. For more on
AHA statements and guidelines development, visit http://professional.heart.org/statements. Select the Guidelines &
Statements drop-down menu, then click Publication Development.

Permissions: Multiple copies, modification, alteration, enhancement, and/or distribution of this document are not
permitted without the express permission of the American Heart Association. Instructions for obtaining permission are
located at http://www.heart.org/HEARTORG/General/Copyright-Permission-Guidelines_UCM_300404_Article.jsp. A
link to the Copyright Permissions Request Form appears on the right side of the page.

(Circulation. 2017;000:e000e000. DOI: 10.1161/CIR.0000000000000548.)

2017 by the American College of Cardiology Foundation, the American Heart Association, Inc., and the Heart Rhythm
Society.

Circulation is available at http://circ.ahajournals.org

Page 2
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table of Contents
Preamble........................................................................................................................................ 5
1. Introduction ................................................................................................................................ 9
1.1. Methodology and Evidence Review ........................................................................................... 9
1.2. Organization of the Writing Committee ................................................................................... 10
1.3. Document Review and Approval ............................................................................................. 10
1.4. Scope of the Guideline .......................................................................................................... 10
1.5. Abbreviations ...................................................................................................................... 15
2. Epidemiology............................................................................................................................. 15
2.1. General Concepts ................................................................................................................. 15
2.1.1. Premature Ventricular Complexes and Nonsustained VT ...................................................... 17
2.1.2. VT and VF During ACS...................................................................................................... 18
2.1.3. Sustained VT and VF Not Associated With ACS .................................................................... 18
2.2. Sudden Cardiac Death ........................................................................................................... 19
2.2.1. Incidence of SCD ............................................................................................................ 19
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

2.2.2. Population Subgroups and Risk Prediction.......................................................................... 19


3. General Evaluation of Patients With Documented or Suspected VA ................................................. 23
3.1. History and Physical Examination............................................................................................ 23
3.2. Noninvasive Evaluation ......................................................................................................... 25
3.2.1. 12-lead ECG and Exercise Testing...................................................................................... 25
3.2.2. Ambulatory Electrocardiography ...................................................................................... 26
3.2.3. Implanted Cardiac Monitors............................................................................................. 26
3.2.4. Noninvasive Cardiac Imaging............................................................................................ 27
3.2.5. Biomarkers.................................................................................................................... 27
3.2.6. Genetic Considerations in Arrhythmia Syndromes ............................................................... 27
3.3. Invasive Testing.................................................................................................................... 28
3.3.1. Invasive Cardiac Imaging: Cardiac Catheterization or CT Angiography..................................... 28
3.3.2. Electrophysiological Study for VA...................................................................................... 28
4. Therapies for Treatment or Prevention of VA ................................................................................ 29
4.1. Medication Therapy.............................................................................................................. 29
4.2. Preventing SCD With HF Medications ...................................................................................... 33
4.3. Surgery and Revascularization Procedures in Patients With Ischemic Heart Disease ....................... 34
4.3.1. Surgery for Arrhythmia Management ................................................................................ 34
4.4. Autonomic Modulation ......................................................................................................... 35
5. Acute Management of Specific VA ............................................................................................... 36
6. Ongoing Management of VA and SCD Risk Related to Specific Disease States .................................... 39
6.1. Ischemic Heart Disease.......................................................................................................... 39
6.1.1. Secondary Prevention of SCD in Patients With Ischemic Heart Disease ................................... 39
6.1.2. Primary Prevention of SCD in Patients With Ischemic Heart Disease....................................... 42
6.1.3. Treatment and Prevention of Recurrent VA in Patients With Ischemic Heart Disease................ 44
6.2. Nonischemic Cardiomyopathy ................................................................................................ 46
6.2.1. Secondary Prevention of SCD in Patients With NICM ........................................................... 47
6.2.2. Primary Prevention of SCD in Patients With NICM ............................................................... 48
6.2.3. Treatment of Recurrent VA in Patients With NICM .............................................................. 49
6.3 Arrhythmogenic Right Ventricular Cardiomyopathy .................................................................... 51
6.4. Hypertrophic Cardiomyopathy................................................................................................ 54
6.5. Myocarditis ......................................................................................................................... 58
6.6. Cardiac Sarcoidosis ............................................................................................................... 58
6.7. Heart Failure........................................................................................................................ 60

Page 3
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.7.1. HF With Reduced Ejection Fraction ................................................................................... 60


6.7.2. Left Ventricular Assist Device ........................................................................................... 60
6.7.3. ICD Use After Heart Transplantation.................................................................................. 61
6.8. Neuromuscular Disorders ...................................................................................................... 61
6.9. Cardiac Channelopathies ....................................................................................................... 63
6.9.1. Specific Cardiac Channelopathy Syndromes........................................................................ 64
7. VA in the Structurally Normal Heart ............................................................................................. 73
7.1. Outflow Tract and Atrioventricular Annular VA ......................................................................... 73
7.2. Papillary Muscle VA .............................................................................................................. 75
7.3. Interfascicular Reentrant VT (Belhassen Tachycardia) ................................................................ 75
7.4. Idiopathic Polymorphic VT/VF ................................................................................................ 76
8. PVC-Induced Cardiomyopathy ..................................................................................................... 77
9. VA and SCD Related to Specific Populations .................................................................................. 77
9.1. Pregnancy ........................................................................................................................... 77
9.2. Older Patients With Comorbidities .......................................................................................... 78
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

9.3. Medication-Induced Arrhythmias............................................................................................ 79


9.4. Adult Congenital Heart Disease............................................................................................... 80
10. Defibrillators Other than Transvenous ICDs ................................................................................. 84
10.1. Subcutaneous Implantable Cardioverter-Defibrillator............................................................... 84
10.2. Wearable Cardioverter-Defibrillator ...................................................................................... 85
11. Special Considerations for Catheter Ablation ............................................................................... 85
12. Postmortem Evaluation of SCD .................................................................................................. 87
13. Terminal Care .......................................................................................................................... 87
14. Shared Decision-Making............................................................................................................ 88
15. Cost and Value Considerations ................................................................................................... 88
16. Quality of Life .......................................................................................................................... 91
17. Evidence Gaps and Future Research Needs .................................................................................. 91
Appendix 1. Author Relationships With Industry and Other Entities (Relevant)..................................... 93
Appendix 2. Reviewer Relationships With Industry and Other Entities (Comprehensive)........................ 96

Page 4
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Preamble
Since 1980, the American College of Cardiology (ACC) and American Heart Association (AHA) have translated
scientific evidence into clinical practice guidelines with recommendations to improve cardiovascular health.
These guidelines, which are based on systematic methods to evaluate and classify evidence, provide a
cornerstone for quality cardiovascular care. The ACC and AHA sponsor the development and publication of
guidelines without commercial support, and members of each organization volunteer their time to the
writing and review efforts. Guidelines are official policy of the ACC and AHA.

Intended Use
Practice guidelines provide recommendations applicable to patients with or at risk of developing
cardiovascular disease. The focus is on medical practice in the United States, but guidelines developed in
collaboration with other organizations may have a global impact. Although guidelines may be used to inform
regulatory or payer decisions, their intent is to improve patients quality of care and align with patients
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

interests. Guidelines are intended to define practices meeting the needs of patients in most, but not all,
circumstances and should not replace clinical judgment.

Clinical Implementation
Guideline-recommended management is effective only when followed by healthcare providers and patients.
Adherence to recommendations can be enhanced by shared decision-making between healthcare providers
and patients, with patient engagement in selecting interventions based on individual values, preferences,
and associated conditions and comorbidities.

Methodology and Modernization


The ACC/AHA Task Force on Clinical Practice Guidelines (Task Force) continuously reviews, updates, and
modifies guideline methodology on the basis of published standards from organizations including the
Institute of Medicine (1, 2) and on the basis of internal reevaluation. Similarly, the presentation and delivery
of guidelines are reevaluated and modified on the basis of evolving technologies and other factors to
facilitate optimal dissemination of information at the point of care to healthcare professionals.
Toward this goal, this guideline heralds the evolved format of presenting guideline recommendations and
associated text called modular knowledge chunk format. Each modular chunk includes a table of related
recommendations, a brief synopsis, recommendation-specific supportive text, and when appropriate, flow
diagrams or additional tables. References are provided within the modular chunk itself to facilitate quick
review. This format also will facilitate seamless updating of guidelines with focused updates as new evidence
is published, and content tagging for rapid electronic retrieval of related recommendations on a topic of
interest. This evolved format was instituted when this guideline was near completion; therefore the current
document represents a transitional formatting that best suits the text as written. Future guidelines will fully
implement this format, including provisions for limiting the amount of text in a guideline.
Recognizing the importance of costvalue considerations in certain guidelines, when appropriate
and feasible, an analysis of the value of a medication, device, or intervention may be performed in
accordance with the ACC/AHA methodology (3).
To ensure that guideline recommendations remain current, new data are reviewed on an ongoing
basis, with full guideline revisions commissioned in approximately 6-year cycles. Publication of new,
potentially practice-changing study results that are relevant to an existing or new medication, device, or

Page 5
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

management strategy will prompt evaluation by the Task Force, in consultation with the relevant guideline
writing committee, to determine whether a focused update should be commissioned. For additional
information and policies regarding guideline development, we encourage readers to consult the ACC/AHA
guideline methodology manual (4) and other methodology articles (5-8).

Selection of Writing Committee Members


The Task Force strives to avoid bias by selecting experts from a broad array of backgrounds. Writing
committee members represent different geographic regions, sexes, ethnicities, races, intellectual
perspectives/biases, and scopes of clinical practice. The Task Force may also invite organizations and
professional societies with related interests and expertise to participate as partners, collaborators, or
endorsers.

Relationships With Industry and Other Entities


The ACC and AHA have rigorous policies and methods to ensure that guidelines are developed without bias
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

or improper influence. The complete relationships with industry and other entities (RWI) policy can be found
online http://www.acc.org/guidelines/about-guidelines-and-clinical-documents/relationships-with-industry-
policy. Appendix 1 of the current document lists writing committee members relevant RWI. For the
purposes of full transparency, writing committee members comprehensive disclosure information is
available online (http://circ.ahajournals.org/lookup/suppl/doi:10.1161/CIR.0000000000000548/-/DC1), as is
the comprehensive disclosure information for the Task Force http://www.acc.org/guidelines/about-
guidelines-and-clinical-documents/guidelines-and-documents-task-forces.

Evidence Review and Evidence Review Committees


When developing recommendations, the writing committee uses evidence-based methodologies that are
based on all available data (4-7). Literature searches focus on randomized controlled trials (RCTs) but also
include registries, nonrandomized comparative and descriptive studies, case series, cohort studies,
systematic reviews, and expert opinion. Only key references are cited.
An independent evidence review committee (ERC) is commissioned when there are 1 questions
deemed of utmost clinical importance that merit formal systematic review. This systematic review will strive
to determine which patients are most likely to benefit from a test, medication, device, or treatment strategy
and to what degree. Criteria for commissioning an ERC and formal systematic review include: a) the absence
of a current authoritative systematic review; b) the feasibility of defining the benefit and risk in a time frame
consistent with the writing of a guideline; c) the relevance to a substantial number of patients; and d) the
likelihood that the findings can be translated into actionable recommendations. ERC members may include
methodologists, epidemiologists, healthcare providers, and biostatisticians. When a formal systematic
review has been commissioned, the recommendations developed by the writing committee on the basis of
the systematic review are marked with SR.

Guideline-Directed Management and Therapy


The term guideline-directed management and therapy (GDMT) encompasses clinical evaluation, diagnostic
testing, and pharmacological and procedural treatments. For these and all recommended medication
treatment regimens, the reader should confirm the dosage by reviewing product insert material and

Page 6
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

evaluate the treatment regimen for contraindications and interactions. The recommendations are limited to
medications, devices, and treatments approved for clinical use in the United States.

Class of Recommendation and Level of Evidence


The Class of Recommendation (COR) indicates the strength of the recommendation, encompassing the
estimated magnitude and certainty of benefit in proportion to risk. The Level of Evidence (LOE) rates the
quality of scientific evidence that supports the intervention on the basis of the type, quantity, and
consistency of data from clinical trials and other sources (Table 1) (4, 6, 8).

The reader is encouraged to consult the full-text guideline (9) for additional guidance and details about the
management of patients with ventricular arrhythmias and the prevention of sudden cardiac death. The
executive summary contains mainly the recommendations.

Glenn N. Levine, MD, FACC, FAHA


Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Chair, ACC/AHA Task Force on Clinical Practice Guidelines

References
1. Committee on Standards for Developing Trustworthy Clinical Practice Guidelines, Institute of Medicine (U.S.).
Clinical Practice Guidelines We Can Trust. Washington, DC: National Academies Press; 2011.
2. Committee on Standards for Systematic Reviews of Comparative Effectiveness Research, Institute of Medicine
(U.S.). Finding What Works in Health Care: Standards for Systematic Reviews. Washington, DC: National Academies
Press; 2011.
3. Anderson JL, Heidenreich PA, Barnett PG, et al. ACC/AHA statement on cost/value methodology in clinical practice
guidelines and performance measures: a report of the American College of Cardiology/American Heart Association
Task Force on Performance Measures and Task Force on Practice Guidelines. Circulation. 2014;129:2329-45.
4. ACCF/AHA Task Force on Practice Guidelines. Methodology Manual and Policies From the ACCF/AHA Task Force on
Practice Guidelines. American College of Cardiology and American Heart Association, 2010. Available at:
http://assets.cardiosource.com/Methodology_Manual_for_ACC_AHA_Writing_Committees.pdf and
http://professional.heart.org/idc/groups/ahamah-
public/@wcm/@sop/documents/downloadable/ucm_319826.pdf. Accessed October 1, 2017.
5. Arnett DK, Goodman RA, Halperin JL, et al. AHA/ACC/HHS strategies to enhance application of clinical practice
guidelines in patients with cardiovascular disease and comorbid conditions: from the American Heart Association,
American College of Cardiology, and U.S. Department of Health and Human Services. Circulation. 2014;130:1662-7.
6. Halperin JL, Levine GN, Al-Khatib SM, et al. Further evolution of the ACC/AHA clinical practice guideline
recommendation classification system: a report of the American College of Cardiology Foundation/American Heart
Association Task Force on Clinical Practice Guidelines. Circulation. 2016;133:1426-8.
7. Jacobs AK, Anderson JL Halperin JL. The evolution and future of ACC/AHA clinical practice guidelines: a 30-year
journey: a report of the American College of Cardiology/American Heart Association Task Force on Practice
Guidelines. Circulation. 2014;130:1208-17.
8. Jacobs AK, Kushner FG, Ettinger SM, et al. ACCF/AHA clinical practice guideline methodology summit report: a
report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice
Guidelines. Circulation. 2013;127:268-310.
9. Al-Khatib SM, Stevenson WG, Ackerman MJ, et al. 2017 AHA/ACC/HRS guideline for management of patients with
ventricular arrhythmias and the prevention of sudden cardiac death: a report of the American College of Cardiology
Foundation/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society
[published online ahead of print October 30, 2017]. Circulation. doi: 10.1161/CIR.0000000000000549.

Page 7
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 1. Applying Class of Recommendation and Level of Evidence to Clinical Strategies, Interventions,
Treatments, or Diagnostic Testing in Patient Care* (Updated August 2015)
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Page 8
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

1. Introduction
1.1. Methodology and Evidence Review
The recommendations listed in this clinical practice guideline are, whenever possible, evidence-based. An
initial extensive evidence review, which included literature derived from research involving human subjects,
published in English, and indexed in MEDLINE (through PubMed), EMBASE, the Cochrane Library, the Agency
for Healthcare Research and Quality, and other selected databases relevant to this guideline, was conducted
from April 2016 to September 2016. Key search words included, but were not limited, to the following: sudden
cardiac death, ventricular tachycardia, ventricular fibrillation, premature ventricular contractions, implantable
cardioverter-defibrillator, subcutaneous implantable cardioverter-defibrillator, wearable cardioverter-
defibrillator, and catheter ablation. Additional relevant studies published through March 2017, during the
guideline writing process, were also considered by the writing committee, and added to the evidence tables
when appropriate. The final evidence tables are included in the Online Data Supplement
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

(http://circ.ahajournals.org/lookup/suppl/doi:10.1161/CIR.0000000000000548/-/DC2) and summarize the


evidence used by the writing committee to formulate recommendations. Additionally, the writing committee
reviewed documents related to ventricular arrhythmias (VA) and sudden cardiac death (SCD) previously
published by the ACC, AHA, and the Heart Rhythm Society (HRS). References selected and published in this
document are representative and not all-inclusive.
As noted in the Preamble, an independent ERC was commissioned to perform a formal systematic
review of 2 important clinical questions for which clear literature and prior guideline consensus were felt to
be lacking or limited (Table 2). The results of the ERC review were considered by the writing committee for
incorporation into this guideline. Concurrent with this process, writing committee members evaluated other
published data relevant to the guideline. The findings of the ERC and the writing committee members were
formally presented and discussed, then guideline recommendations were developed. The Systematic Review
for the 2017 AHA/ACC/HRS Guideline for Management of Patients With Ventricular Arrhythmias and the
Prevention of Sudden Cardiac Death is published in conjunction with this guideline (1).

Table 2. Systematic Review Questions on SCD Prevention


Question
Question Section Number
Number
For asymptomatic patients with Brugada syndrome, what is the association
1 between an abnormal programmed ventricular stimulation study and SCD and 7.9.1.3
other arrhythmia endpoints?
What is the impact of ICD implantation for primary prevention in older patients
2 9.3
and patients with significant comorbidities?
ICD indicates implantable cardioverter-defibrillator; and SCD, sudden cardiac death.

The ACC and AHA have acknowledged the importance of value in health care and have called for eventual
development of a Level of Value for clinical practice recommendations (2). Available cost-effectiveness data
were determined to be sufficient to support 2 specific recommendations in this guideline (see Sections 7.1.1
and 7.1.2). As a result, a Level of Value was assigned to those 2 recommendations on the basis of the
ACC/AHA Statement on Cost/Value Methodology in Clinical Practice Guidelines and Performance Measures,
as shown in Table 3 (2). Available quality of life (QoL) data were deemed to be insufficient to support specific
recommendations in this guideline.

Page 9
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 3. Proposed Integration of Level of Value Into Clinical Practice Guideline Recommendations*
Level of Value
High value: Better outcomes at lower cost or ICER <$50,000 per QALY gained
Intermediate value: $50,000 to <$150,000 per QALY gained
Low value: $150,000 per QALY gained
Uncertain value: Value examined but data are insufficient to draw a conclusion because of no
studies, low-quality studies, conflicting studies, or prior studies that are no longer relevant
Not assessed: Value not assessed by the writing committee
Proposed abbreviations for each value recommendation:
Level of Value: H to indicate high value; I, intermediate value; L, low value; U, uncertain value; and
NA, value not assessed
*Dollar amounts used in this table are based on U.S. GDP data from 2012 and were obtained from WHO-CHOICE Cost-
Effectiveness Thresholds (3).
GDP indicates gross domestic product; ICER, incremental cost-effectiveness ratio; QALY, quality-adjusted life-years; and
WHO-CHOICE, World Health Organization Choosing Interventions that are Cost-Effective.
Reproduced from Anderson, et al. (2).
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

1.2. Organization of the Writing Committee


The writing committee consisted of cardiac electrophysiologists (including those specialized in pediatrics),
general adult and pediatric cardiologists (including those specialized in critical care and acute coronary
syndromes [ACS], genetic cardiology, heart failure, and cost-effectiveness analyses), a geriatrician with
expertise in terminal care and shared decision-making, and a lay representative, in addition to representatives
from the ACC, AHA, HRS, and the Heart Failure Society of America (HFSA).

1.3. Document Review and Approval


This document was reviewed by 2 official reviewers nominated by the ACC, AHA, and HRS; 1 official lay
reviewer nominated by the AHA; 1 organizational reviewer nominated by the HFSA; and 28 individual content
reviewers. Reviewers RWI information was distributed to the writing committee and is published in this
document (Appendix 2).
This document was approved for publication by the governing bodies of the ACC, the AHA, and the
HRS; and endorsed by the HFSA.

1.4. Scope of the Guideline


The purpose of this AHA/ACC/HRS document is to provide a contemporary guideline for the management of
adults who have VA or who are at risk for SCD, including diseases and syndromes associated with a risk of SCD
from VA. This guideline supersedes the ACC/AHA/ESC 2006 Guidelines for Management of Patients With
Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death (4). It also supersedes some sections of
the ACC/AHA/HRS 2008 Guidelines for Device-Based Therapy of Cardiac Rhythm Abnormalities (5),
specifically those sections on indications for the implantable cardioverter-defibrillator (ICD); and, it updates
the SCD prevention recommendations in the 2011 ACCF/AHA Guideline for the Diagnosis and Treatment of
Hypertrophic Cardiomyopathy (6). Some recommendations from the earlier guidelines have been updated
as warranted by new evidence or a better understanding of existing evidence, and irrelevant or overlapping
recommendations were deleted or modified.
In the current guideline, sudden cardiac arrest (SCA) is defined as the sudden cessation of cardiac
activity so that the victim becomes unresponsive, with no normal breathing and no signs of circulation (7). If
corrective measures are not taken rapidly, this condition progresses to SCD. Cardiac arrest is used to signify
an event that can be reversed, usually by cardiopulmonary resuscitation (CPR), administration of medications

Page 10
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

and/or defibrillation or cardioversion. SCA and SCD can result from causes other than VA, such as
bradyarrhythmias, electromechanical dissociation, pulmonary embolism, intracranial hemorrhage, and aortic
dissection; however, the scope of this document includes only SCA and SCD due to VA.
This guideline includes indications for ICDs for the treatment of VA and prevention of SCD, but it does
not delve into details on individual device selection and programming, including considerations relevant to
cardiac resynchronization therapy (CRT), bradycardia pacing, and hemodynamic monitoring. These important
aspects of ICD management have been covered in an HRS expert consensus statement (8). An AHA science
advisory discusses the use of wearable cardioverter-defibrillators (9). The findings of that document were
reviewed; however, recommendations on this topic were developed independently of that document. This
guideline includes indications for catheter ablation of VA, but does not provide recommendations on specific
techniques or ablation technologies, which were beyond the scope of this document.
Recommendations for interventional therapies, including ablation and the implantation of devices,
apply only if these therapies can be implemented by qualified clinicians, such that outcomes consistent with
published literature are a reasonable expectation. The writing committee agreed that a high degree of
expertise was particularly important for performance of catheter ablation of VA, and this point is further
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

emphasized in relevant sections. In addition, all recommendations related to ICDs require that meaningful
survival of >1 year is expected; meaningful survival means that a patient has a reasonable quality of life and
functional status.
Although this document is aimed at the adult population (18 years of age) and offers no specific
recommendations for pediatric patients, some of the literature on pediatric patients was examined. In some
cases, the data from pediatric patients beyond infancy helped to inform this guideline.
The writing committee recognized the importance of shared decision-making and patient-centered care and,
when possible, it endeavored to formulate recommendations relevant to these important concepts. The
importance of a shared decision-making process in which the patient, family, and clinicians discuss risks and
benefits of diagnostic and treatment options and consider the patients personal preferences is emphasized
(see Section 15).
In developing this guideline, the writing committee reviewed previously published guidelines and
related statements. Table 4 contains a list of guidelines and statements deemed pertinent to this writing effort
and is intended for use as a resource, obviating repetition of existing guideline recommendations.

Page 11
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 4. Associated Guidelines and Statements


Publication Year
Title Organization
(Reference)
Guidelines
Syncope ACC/AHA/HRS 2017 (10)
2017 (11) 2016 (12), and
Heart failure ACCF/AHA
2013 (13)
Valvular heart disease AHA/ACC 2017 (14) and 2014 (15)
Supraventricular tachycardia ACC/AHA/HRS 2015 (16)
Ventricular arrhythmias and the prevention of sudden cardiac
ESC 2015 (17)
death
Guidelines for cardiopulmonary resuscitation and emergency
AHA 2015 (18)
cardiovascular care
Atrial fibrillation AHA/ACC/HRS 2014 (19)
NonST-elevation acute coronary syndromes AHA/ACC 2014 (20)
Assessment of cardiovascular risk ACC/AHA 2013 (21)
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

ST-elevation myocardial infarction ACCF/AHA 2013 (22)


Acute myocardial infarction in patients presenting with ST-
ESC 2012 (23)
segment elevation
Device-based therapies for cardiac rhythm abnormalities ACCF/AHA/HRS 2012 (24)
Coronary artery bypass graft surgery ACCF/AHA 2011 (25)
Hypertrophic cardiomyopathy ACCF/AHA 2011 (6)
Percutaneous coronary intervention ACCF/AHA/SCAI 2011 (26)
Secondary prevention and risk reduction therapy for patients
AHA/ACCF 2011 (27)
with coronary and other atherosclerotic vascular disease
Scientific Statements
Wearable cardioverter-defibrillator therapy for the prevention
AHA 2016 (9)
of sudden cardiac death
Optimal implantable cardioverter defibrillator programming and HRS/EHRA/APHRS/
2016 (8)
testing SOLAECE
Treatment of cardiac arrest: current status and future directions:
IOM 2015 (28)
strategies to improve cardiac arrest survival
Eligibility and disqualification recommendations for competitive
ACC/AHA 2015 (29)
athletes with cardiovascular abnormalities
Ventricular arrhythmias EHRA/HRS/APHRS 2014 (30)
Arrhythmias in adult congenital heart disease PACES/HRS 2014 (31)
Implantable cardioverter-defibrillator therapy in patients who
HRS/ACC/AHA 2014 (32)
are not included or not well represented in clinical trials
Cardiac sarcoidosis HRS 2014 (33)
Inherited primary arrhythmia syndromes HRS/EHRA/APHRS 2013 (34)
ACC indicates American College of Cardiology; ACCF, American College of Cardiology Foundation; AHA, American Heart
Association; APHRS, Asia Pacific Heart Rhythm Society; EHRA, European Heart Rhythm Association; ESC, European Society
of Cardiology; HRS, Heart Rhythm Society; PACES, Pediatric and Congenital Electrophysiology Society; SCAI, Society for
Cardiovascular Angiography and Interventions; and, SOLAECE, Sociedad Latinoamericana de Estimulacion Cardiaca y
Electrofisiologia.

References
1. Kusumoto FM, Bailey KR, Chaouki AS, et al. Systematic review for the 2017 AHA/ACC/HRS guideline for management
of patients with ventricular arrhythmias and the prevention of sudden cardiac death: a report of the American

Page 12
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm
Society [published online ahead of print October 30, 2017]. Circulation. doi: 10.1161/CIR.0000000000000550.
2. Anderson JL, Heidenreich PA, Barnett PG, et al. ACC/AHA statement on cost/value methodology in clinical practice
guidelines and performance measures: a report of the American College of Cardiology/American Heart Association
Task Force on Performance Measures and Task Force on Practice Guidelines. Circulation. 2014;129:2329-45.
3. World Health Organization. CHOosing Interventions that are Cost Effective (WHO-CHOICE): cost-effectiveness
thresholds. Available at: http://www.who.int/choice/en/. Accessed March 26, 2013.
4. Zipes DP, Camm AJ, Borggrefe M, et al. ACC/AHA/ESC 2006 guidelines for management of patients with ventricular
arrhythmias and the prevention of sudden cardiac death: a report of the American College of Cardiology/American
Heart Association Task Force and the European Society of Cardiology Committee for Practice Guidelines (Writing
Committee to Develop Guidelines for Management of Patients With Ventricular Arrhythmias and the Prevention of
Sudden Cardiac Death). Developed in collaboration with the European Heart Rhythm Association and the Heart
Rhythm Society. Circulation. 2006;114:e385-484.
5. Epstein AE, DiMarco JP, Ellenbogen KA, et al. ACC/AHA/HRS 2008 guidelines for device-based therapy of cardiac
rhythm abnormalities: a report of the American College of Cardiology/American Heart Association Task Force on
Practice Guidelines (Writing Committee to Revise the ACC/AHA/NASPE 2002 Guideline Update for Implantation of
Cardiac Pacemakers and Antiarrhythmia Devices). Developed in collaboration with the American Association for
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Thoracic Surgery and Society of Thoracic Surgeons. Circulation. 2008;117:e350-408.


6. Gersh BJ, Maron BJ, Bonow RO, et al. 2011 ACCF/AHA guideline for the diagnosis and treatment of hypertrophic
cardiomyopathy: a report of the American College of Cardiology Foundation/American Heart Association Task Force
on Practice Guidelines. Developed in collaboration with the American Association for Thoracic Surgery, American
Society of Echocardiography, American Society of Nuclear Cardiology, Heart Failure Society of America, Heart
Rhythm Society, Society for Cardiovascular Angiography and Interventions, and Society of Thoracic Surgeons.
Circulation. 2011;124:783-831.
7. Buxton AE, Calkins H, Callans DJ, et al. ACC/AHA/HRS 2006 key data elements and definitions for
electrophysiological studies and procedures: a report of the American College of Cardiology/American Heart
Association Task Force on Clinical Data Standards (ACC/AHA/HRS Writing Committee to Develop Data Standards on
Electrophysiology). Circulation. 2006;114:2534-70.
8. Wilkoff BL, Fauchier L, Stiles MK, et al. 2015 HRS/EHRA/APHRS/SOLAECE expert consensus statement on optimal
implantable cardioverter-defibrillator programming and testing. J Arrhythm. 2016;32:1-28.
9. Piccini JP Sr, Allen LA, Kudenchuk PJ, et al. Wearable cardioverter-defibrillator therapy for the prevention of sudden
cardiac death: a science advisory from the American Heart Association. Circulation. 2016;133:1715-27.
10.Shen WK, Sheldon RS, Benditt DG, et al. 2017 ACC/AHA/HRS guideline for the evaluation and management of
patients with syncope: a report of the American College of Cardiology/American Heart Association Task Force on
Clinical Practice Guidelines and the Heart Rhythm Society. Circulation. 2017;136:e60-122.
11.Yancy CW, Jessup M, Bozkurt B, et al. 2017 ACC/AHA/HFSA focused update of the 2013 ACCF/AHA guideline for the
management of heart failure: a report of the American College of Cardiology/American Heart Association Task Force
on Clinical Practice Guidelines and the Heart Failure Society of America. Circulation. 2017;136:e137-61.
12.Yancy CW, Jessup M, Bozkurt B, et al. 2016 ACC/AHA/HFSA focused update on new pharmacological therapy for
heart failure: an update of the 2013 ACCF/AHA guideline for the management of heart failure: a report of the
American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart
Failure Society of America. Circulation. 2016;134:e282-93.
13.Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for the management of heart failure: a report of the
American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines.
Circulation. 2013;128:e240-327.
14.Nishimura RA, Otto CM, Bonow RO, et al. 2017 AHA/ACC focused update of the 2014 AHA/ACC guideline for the
management of patients with valvular heart disease: a report of the American College of Cardiology/American Heart
Association Task Force on Clinical Practice Guidelines. Circulation. 2017;135:e1159-95.
15.Nishimura RA, Otto CM, Bonow RO, et al. 2014 AHA/ACC guideline for the management of patients with valvular
heart disease: a report of the American College of Cardiology/American Heart Association Task Force on Practice
Guidelines. Circulation. 2014;129:e521-643.
16.Page RL, Joglar JA, Caldwell MA, et al. 2015 ACC/AHA/HRS guideline for the management of adult patients with
supraventricular tachycardia: a report of the American College of Cardiology/American Heart Association Task Force
on Clinical Practice Guidelines and the Heart Rhythm Society. Circulation. 2016;133:e506-74.

Page 13
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

17.Priori SG, Blomstrom-Lundqvist C, Mazzanti A, et al. 2015 ESC guidelines for the management of patients with
ventricular arrhythmias and the prevention of sudden cardiac death. The Task Force for the Management of
Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death of the European Society of
Cardiology. G Ital Cardiol (Rome). 2016;17:108-70.
18.Perkins GD, Jacobs IG, Nadkarni VM, et al. Cardiac arrest and cardiopulmonary resuscitation outcome reports:
update of the Utstein Resuscitation Registry Templates for Out-of-Hospital Cardiac Arrest: a statement for
healthcare professionals from a task force of the International Liaison Committee on Resuscitation (American Heart
Association, European Resuscitation Council, Australian and New Zealand Council on Resuscitation, Heart and Stroke
Foundation of Canada, InterAmerican Heart Foundation, Resuscitation Council of Southern Africa, Resuscitation
Council of Asia);and the American Heart Association Emergency Cardiovascular Care Committee and the Council on
Cardiopulmonary, Critical Care, Perioperative and Resuscitation. Circulation. 2015;132:1286-300.
19.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. Circulation. 2014;130:e199-267.
20.Amsterdam EA, Wenger NK, Brindis RG, et al. 2014 AHA/ACC 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:e344-426.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

21.Andrus B, Lacaille D. 2013 ACC/AHA guideline on the assessment of cardiovascular risk. J Am Coll Cardiol.
2014;63:2886.
22.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.
23.Steg PG, James SK, Atar D, et al. ESC guidelines for the management of acute myocardial infarction in patients
presenting with ST-segment elevation. Eur Heart J. 2012;33:2569-619.
24.Tracy CM, Epstein AE, Darbar D, et al. 2012 ACCF/AHA/HRS focused update of the 2008 guidelines for device-based
therapy of cardiac rhythm abnormalities: a report of the American College of Cardiology Foundation/American
Heart Association Task Force on Practice Guidelines. Circulation. 2012;126:1784-800.
25.Hillis LD, Smith PK, Anderson JL, et al. 2011 ACCF/AHA guideline for coronary artery bypass graft surgery: a report of
the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines.
Developed in collaboration with the American Association for Thoracic Surgery, Society of Cardiovascular
Anesthesiologists, and Society of Thoracic Surgeons. Circulation. 2011;124:e652-735.
26.Levine GN, Bates ER, Blankenship JC, et al. 2011 ACCF/AHA/SCAI guideline for percutaneous coronary intervention: a
report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice
Guidelines and the Society for Cardiovascular Angiography and Interventions. Circulation. 2011;124:e574-651.
27.Smith SC Jr, Benjamin EJ, Bonow RO, et al. AHA/ACCF secondary prevention and risk reduction therapy for patients
with coronary and other atherosclerotic vascular disease: 2011 update: a guideline from the American Heart
Association and American College of Cardiology Foundation. Circulation. 2011;124:2458-73.
28.Institute of Medicine. Committee on the Treatment of Cardiac Arrest: Current status and future directions:
strategies to improve cardiac arrest survival: a time to act. Washington, DC: National Academic Press; 2015.
29.Link MS, Myerburg RJ, Estes NA 3rd. Eligibility and disqualification recommendations for competitive athletes with
cardiovascular abnormalities: Task Force 12: emergency action plans, resuscitation, cardiopulmonary resuscitation,
and automated external defibrillators: a scientific statement from the American Heart Association and American
College of Cardiology. Circulation. 2015;132:e334-8.
30.Pedersen CT, Kay GN, Kalman J, et al. EHRA/HRS/APHRS expert consensus on ventricular arrhythmias. Heart
Rhythm. 2014;11:e166-96.
31.Khairy P, Van Hare GF, Balaji S, et al. PACES/HRS expert consensus statement on the recognition and management
of arrhythmias in adult congenital heart disease: developed in partnership between the Pediatric and Congenital
Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Heart Rhythm. 2014;11:e102-65.
32.Kusumoto FM, Calkins H, Boehmer J, et al. HRS/ACC/AHA expert consensus statement on the use of implantable
cardioverter-defibrillator therapy in patients who are not included or not well represented in clinical trials.
Circulation. 2014;130:94-125.
33.Birnie DH, Sauer WH, Bogun F, et al. HRS expert consensus statement on the diagnosis and management of
arrhythmias associated with cardiac sarcoidosis. Heart Rhythm. 2014;11:1305-23.

Page 14
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

34.Priori SG, Wilde AA, Horie M, et al. HRS/EHRA/APHRS expert consensus statement on the diagnosis and
management of patients with inherited primary arrhythmia syndromes. Heart Rhythm. 2013;10:1932-63.

1.5. Abbreviations
Abbreviation Meaning/Phrase
ACS acute coronary syndrome
CPR cardiopulmonary resuscitation
CRT cardiac resynchronization therapy
ECG electrocardiogram
ERC evidence review committee
GDMT guideline-directed management and therapy
HCM hypertrophic cardiomyopathy
HF heart failure
HFpEF heart failure with preserved ejection fraction
HFrEF heart failure with reduced ejection fraction
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

ICD implantable cardioverter-defibrillator


LV left ventricular
LVAD left ventricular assist device
LVEF left ventricular ejection fraction
MI myocardial infarction
NICM nonischemic cardiomyopathy
NSVT nonsustained ventricular tachycardia
PCI percutaneous coronary intervention
PVC premature ventricular complex
QoL quality of life
RCT randomized controlled trial
RVOT right ventricular outflow tract
SCA sudden cardiac arrest
SCD sudden cardiac death
VA ventricular arrhythmia
VT ventricular tachycardia

2. Epidemiology
2.1. General Concepts
Table 5
VA include a spectrum that ranges from premature ventricular complex (PVC) to ventricular fibrillation (VF),
with a clinical presentation that ranges from a total lack of symptoms to cardiac arrest. Most life-threatening
VA are associated with ischemic heart disease, particularly in older patients (1). The risks of VA and SCD vary
in specific populations with different underlying cardiac conditions, and with specific family history and genetic
variants, and this variation has important implications for studying and applying therapies.

Page 15
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 5. Table of Definitions of Commonly Used Terms in this Document


Term Definition or Description
Ventricular tachycardia (2) Cardiac arrhythmia of 3 consecutive complexes originating in the ventricles at a rate
>100 bpm (cycle length: <600 ms). Types of VT:
Sustained: VT >30 s or requiring termination due to hemodynamic compromise in
<30 s.
Nonsustained/unsustained: 3 beats, terminating spontaneously.
Monomorphic: Stable single QRS morphology from beat to beat.
Polymorphic: Changing or multiform QRS morphology from beat to beat.
Bidirectional: VT with a beat-to-beat alternation in the QRS frontal plane axis,
often seen in the setting of digitalis toxicity or catecholaminergic polymorphic VT

Monomorphic VT
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Polymorphic VT

Bidirectional VT

Torsades de pointes (2) Torsades de pointes is polymorphic VT that occurs in the setting of a long-QT interval
and is characterized by a waxing and waning QRS amplitude. It often has a long-short
initiating sequence with a long coupling interval to the first VT beat and may present
with salvos of NSVT. The twisting of the points, although characteristic, may not always
be seen, especially if the episode is nonsustained or if only a limited number of leads
are available. Torsades de pointes can result from bradycardia including high-grade AV
block that leads to a long-short sequence initiating torsades de pointes.

Ventricular flutter (2) A regular VA 300 bpm (cycle length: 200 ms) with a sinusoidal, monomorphic
appearance; no isoelectric interval between successive QRS complexes.

Page 16
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Ventricular fibrillation (2) Rapid, grossly irregular electrical activity with marked variability in
electrocardiographic waveform, ventricular rate usually >300 bpm (cycle length: <200
ms).

Sudden cardiac arrest (2) SCA is the sudden cessation of cardiac activity such that the victim becomes
unresponsive, with either persisting gasping respirations or absence of any respiratory
movements, and no signs of circulation as manifest by the absence of a perceptible
pulse. An arrest is presumed to be of cardiac etiology unless it is known or likely to have
been caused by trauma, drowning, respiratory failure or asphyxia, electrocution, drug
overdose, or any other noncardiac cause.
Sudden cardiac death (2) Sudden and unexpected death occurring within an hour of the onset of symptoms, or
occurring in patients found dead within 24 h of being asymptomatic and presumably
due to a cardiac arrhythmia or hemodynamic catastrophe.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

VT/VF storm (3) VT/VF storm (electrical storm or arrhythmic storm) refers to a state of cardiac electrical
instability that is defined by 3 episodes of sustained VT, VF, or appropriate shocks
from an ICD within 24 h.
Primary prevention ICD (2) ICD placement with the intention of preventing SCD in a patient who has not had
sustained VT or SCA but who is at an increased risk for these events.
Secondary prevention ICD (2) ICD placement in a patient with prior SCA, sustained VT, or syncope caused by VA.
Structural heart disease* TThis term encompasses IHD, all types of cardiomyopathy, valvular heart disease, and
adult congenital heart disease.
Cardiac channelopathy (4) Arrhythmogenic disease due to a genetic abnormality that results in dysfunction of a
cardiac ion channel (e.g., long-QT syndrome, catecholaminergic polymorphic VT).
*The definition of this term may differ across publications. Refer to the entry for the definition used in this document.
AV indicates atrioventricular; ICD, implantable cardioverter-defibrillator; IHD, ischemic heart disease; NSVT,
nonsustained ventricular tachycardia; SCA, sudden cardiac arrest; SCD, sudden cardiac death; VA, ventricular arrhythmia;
VF, ventricular fibrillation; and VT, ventricular tachycardia.

2.1.1. Premature Ventricular Complexes and Nonsustained VT


PVCs are common and increase in frequency with age. Although PVCs were found in a healthy military
population in only 0.6% of those <20 years of age and 2.7% of those >50 years of age (5) on 12-lead ECGs,
longer term monitoring shows PVCs in about 50% of all people with or without heart disease (6). The presence
of PVCs on 2 minutes of monitoring of middle-aged patients in the ARIC (Atherosclerosis Risk In Communities)
study was associated with increased risk of both ischemic heart disease events and mortality, with or without
prevalent ischemic heart disease (7, 8). In the general population, frequent PVCs, which are defined as the
presence of at least 1 PVC on a 12-lead ECG or >30 PVCs per hour, are associated with increased cardiovascular
risk and increased mortality (9). In a study from Taiwan of patients without sustained VT or structural heart
disease who had 24-hour Holter monitoring for clinical evaluation, multifocal PVCs were associated with
increased risk of death and nonfatal cardiovascular adverse outcomes (10). In the same population,
nonsustained ventricular tachycardia (NSVT) was independently associated with increased risk of death and
other cardiovascular adverse outcomes, including stroke (11). An association of PVCs with increased risk of
stroke was also seen in the ARIC population (8).
Because some studies have shown an association of PVCs with adverse outcomes, the detection of
PVCs, particularly if multifocal and frequent, is generally considered a risk factor for adverse cardiovascular
outcomes, and such patients are generally evaluated to ensure they do not have underlying conditions (e.g.,
ischemic heart disease, left ventricular [LV] dysfunction) that warrant further treatment to reduce risk. PVC
and NSVT in patients with cardiovascular disease are common and have been associated with adverse

Page 17
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

outcomes (12, 13). In CAST (Cardiac Arrhythmia Suppression Trials), treatment of patients with post-
myocardial infarction (MI) who took antiarrhythmic medications (e.g., flecainide, encainide, moricizine)
increased the risk of death despite suppression of VA (14, 15). Treatment of PVCs with antiarrhythmic
medications has not been shown to reduce mortality and, in the post- MI population, treatment with class I
sodium channelblocking medications (e.g., quinidine, flecainide) increases the risk of death (15, 16). Likewise,
in patients with a reduced LVEF class I, sodium channelblocking medications and d-sotalol increase the risk
of death (16, 17). Beta blockers, nondihydropyridines calcium channel blockers, and some antiarrhythmic
medications may relieve symptoms of palpitations (18).
PVCs that occur during an exercise test are associated with a higher risk of death (19). In 1 study, PVCs
that occur during recovery are a stronger predictor of death than PVCs occurring only during exercise (20).
However, PVCs are common in trained athletes who have palpitations, in whom there does not appear to be
increased risk of death based on studies of small numbers of athletes, at least in those without other
cardiovascular abnormalities (21, 22). Complex PVCs may not represent a benign finding in endurance
athletes. An electrophysiological study may be needed to assess patients arrhythmogenic risk (22). Very
frequent PVCs, >10,000 to 20,000 a day, can be associated with depressed LV function in some patients that
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

is reversible with control of the PVCs, and has been referred to as PVC-induced cardiomyopathy (23, 24). (See
also Section 8.5. PVC-Induced Cardiomyopathy.) Very rarely, idiopathic PVCs from the outflow tract may
trigger malignant VA in patients without structural heart disease (25, 26).

2.1.2. VT and VF During ACS


Approximately half of patients with out-of-hospital cardiac arrest with the first rhythm identified as VF and
who survive to hospital admission have evidence of acute MI (AMI) (27). Of all out-of-hospital cardiac arrests,
>50% will have significant coronary artery lesions on acute coronary angiography (27). Of patients hospitalized
with AMI, 5% to 10% have VF or sustained VT prior to hospital presentation, and another 5% will have VF or
sustained VT after hospital arrival, most within 48 hours of admission. A study of patients with nonST-
elevation ACS who underwent cardiac catheterization within 48 hours found VT/VF in 7.6% of patients, with
60% of those events within 48 hours of admission (28). Accelerated idioventricular rhythm is a common
arrhythmia in patients with acute MI, including patients with ST-segment elevation MI undergoing primary
percutaneous coronary intervention (PCI). Accelerated idioventricular rhythm is more closely related to the
extent of infarction than to reperfusion itself (29).
Sustained VA that occurs in the setting of an ACS is more often polymorphic VT or VF than
monomorphic VT. Risk factors for VT/VF include prior history of hypertension, prior MI, ST-segment changes
at presentation, and chronic obstructive pulmonary disease (30). A nationwide Danish study found that 11.6%
of patients with ST-segment elevation MI who underwent PCI had VF prior to the PCI, and that VF was
associated with alcohol consumption, preinfarction angina, anterior infarct location, and complete coronary
occlusion at the time of coronary angiography (31). In a select group of patients undergoing primary PCI in a
clinical trial, 5.7% developed sustained VT or VF, with two thirds of these events occurring prior to the end of
the catheterization, and 90% within 48 hours from the procedure. VT or VF after primary PCI was associated
with lower blood pressure, higher heart rate, poor coronary flow at the end of the procedure, and incomplete
resolution of ST elevation (32). Importantly, and in contrast to some earlier studies, VT or VF at any time was
associated with a substantially higher risk of death within 90 days. Late VT or VF (after 48 hours of hospital
presentation) was associated with a higher risk of death than early VT or VF (within 48 hours of hospital
presentation) (33).

2.1.3. Sustained VT and VF Not Associated With ACS


Patients with structural heart disease are at an increased risk for sustained VT and VF. Sustained VT that is not
associated with an ACS is often monomorphic as it is usually due to scar-related reentry, but it may degenerate
to VF (34). The risk and predictors of VT in patients with structural heart disease depend on the type, severity,

Page 18
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

and duration of structural heart disease, increasing with the severity of ventricular dysfunction and the
presence of symptomatic HF. Monomorphic VT occurring in the absence of structural heart disease is
commonly referred to as idiopathic VT and is often due to an automatic focus in a characteristic location,
giving rise to typical electrocardiographic appearances. Polymorphic VT and VF occurring in the absence of
structural heart disease are rare and may be due to a cardiac channelopathy (35, 36), medication-induced long
QT syndrome (36), or they may be idiopathic (37, 38).

2.2. Sudden Cardiac Death


2.2.1. Incidence of SCD
SCA and its most common consequence, SCD, constitute major public health problems, accounting for
approximately 50% of all cardiovascular deaths (1, 39), with at least 25% being first symptomatic cardiac
events (1, 40, 41). In addition, analyses of the magnitude of SCD are limited, in part because of the broad range
of estimates of the risk based on different epidemiological methods (42). During the past 20 to 30 years, SCD
accounted for approximately 230,000 to 350,000 deaths per year in the United States, with a range of
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

<170,000 to >450,000, depending on epidemiological methods, data sources, and inclusion criteria (41, 43).
The lowest of these extremes came from national extrapolation of data from specific local programs, while
the highest rates included noncardiac causes of sudden death such as pulmonary embolism or intracranial
bleeding. The mid-range numbers were largely based on death certificate studies that required a code
inclusive of ischemic heart disease.
The 2017 update of cardiovascular statistics from the AHA estimated the total annual burden of out-
of-hospital cardiac arrest at 356,500 (44). An additional 209,000 in-hospital cardiac arrests occur annually (45).
Among the out-of-hospital cardiac arrest group, approximately 357,000 events trigger emergency rescue
response, with 97% occurring in adults >18 years of age.
The survival statistics for out-of-hospital cardiac arrest remain disappointing, with an estimated 10%
overall survival rate (44). Among the subgroup of 70% of out-of-hospital cardiac arrests that occur in the home,
survival is 6%. The best reported outcomes are from locations with highly developed and publicly visible
emergency rescue response, along with the combination of public location of cardiac arrest, bystander
witnesses willing to provide CPR, first responders arriving quickly, shockable rhythm at initial contact,
availability of automated external defibrillators (AEDs), and possibly a benefit from telecommunication-
directed CPR (46, 47). Survival to hospital discharge after in-hospital cardiac arrests is estimated to be 24%
(48). In all settings, survival statistics appear to be better when rhythms recorded by responders are shockable
(VF, pulseless VT), compared with pulseless electrical activity or asystole (49). Although the apparent increase
in the incidence of pulseless electrical activity or asystole could be due to the later arrival of medical care, the
decrease in the incidence of shockable rhythm has also been attributed, in part, to improvements in diagnosis
and treatment of structural heart disease (40).

2.2.2. Population Subgroups and Risk Prediction


SRisk prediction for SCA and SCD is complex. Risk analysis is divided into 2 general categories: population risk
prediction and individual risk prediction (41, 50). Conventional epidemiological markers provide insight into
probabilities for the development of ischemic heart disease within a general class of subjects, but adequately
tested and validated profiles for SCA risk stratification of individuals in the general population do not presently
exist. The challenge of defining SCA risk in individuals derives from a population model characterized by large
numbers of events diluted into a very large denominator (Figure 1). The overall population can be subgrouped
into categories based on integration of age, presence and extent of disease, and identification of small, high-
risk subgroups within the large denominator general population.

Page 19
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Increasing age is a strong predictor of risk for SCA, but it is not linear. Risk in the general population,
over time, beginning at 35 years of age has been estimated at 1 per 1000 population per year, increasing from
a risk <1000 at the younger end of that spectrum to a higher risk in the elderly (41). However, an analysis of
lifetime risk of SCD, derived from the Framingham data, suggested that the incidence of SCD decreases in later
years, especially in people >75 years of age (51). The data also suggested that SCD is uniformly more common
in men than in women at all age groups. In contrast, the population of children, adolescents, and young adults
has an overall annual risk of 1 per 100,000, and there is somewhat a higher risk of SCD at the younger end of
that age range (41). An age-associated transition range, from the mid-20s to 35 to 40 years of age, is
characterized by a steep increase in risk from that of the adolescent group to the middle-aged group,
corresponding to the emergence of ischemic heart disease.
Although ischemic heart disease remains the most common underlying substrate associated with SCD,
the incidence of ischemic heart disease-related SCD appears to be decreasing (52), with various forms of
cardiomyopathy associated with myocardial fibrosis and LV hypertrophy increasing (53). In addition, a trend
over time has suggested that out-of-hospital cardiac arrest patients who are admitted alive to a hospital are
becoming more likely to have high-risk clinical profiles, as opposed to manifest disease (54). The younger
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

populationchildren, adolescents, and young adultsis affected by a series of disorders that manifest earlier
in life, including the genetic structural disorders and cardiac channelopathies, myocarditis, congenital heart
disease, and other rare disorders (43). During the transition range, from the mid-20s to the mid-30s, causes
of SCA and SCD include a lower proportion of inherited diseases and increasing proportion of ischemic heart
disease (>40% of cases) (43).
Despite the small progress that has been made in risk prediction of SCA and SCD, the greatest
challenge is to identify the relatively small, high-risk subgroups concealed within the large general population
who have no identified disease but are at risk of SCA as their first cardiac event (Figure 1) (50).

Figure 1A. SCD Incidence and Total Events (1)

EF indicates ejection fraction; and SCD, sudden cardiac death.

Page 20
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 1B. SCD and Clinical Subsets (1)


Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

SCD indicates sudden cardiac death.

References
1. Myerburg RJ, Junttila MJ. Sudden cardiac death caused by coronary heart disease. Circulation. 2012;125:1043-52.
2. Buxton AE, Calkins H, Callans DJ, et al. ACC/AHA/HRS 2006 key data elements and definitions for
electrophysiological studies and procedures: a report of the American College of Cardiology/American Heart
Association Task Force on Clinical Data Standards. Circulation. 2006;114:2534-70.
3. Sapp JL, Wells GA, Parkash R, et al. Ventricular tachycardia ablation versus escalation of antiarrhythmic drugs. N Engl
J Med. 2016;375:111-21.
4. Priori SG, Wilde AA, Horie M, et al. HRS/EHRA/APHRS expert consensus statement on the diagnosis and
management of patients with inherited primary arrhythmia syndromes. Heart Rhythm. 2013;10:1932-63.
5. Hiss RG, Lamb LE. Electrocardiographic findings in 122,043 individuals. Circulation. 1962;25:947-61.
6. Brodsky M, Wu D, Denes P, et al. Arrhythmias documented by 24 hour continuous electrocardiographic monitoring
in 50 male medical students without apparent heart disease. Am J Cardiol. 1977;39:390-5.
7. Massing MW, Simpson RJ Jr, Rautaharju PM, et al. Usefulness of ventricular premature complexes to predict
coronary heart disease events and mortality (from the Atherosclerosis Risk In Communities cohort). Am J Cardiol.
2006;98:1609-12.
8. Ofoma U, He F, Shaffer ML, et al. Premature cardiac contractions and risk of incident ischemic stroke. J Am Heart
Assoc. 2012;1:e002519.
9. Ataklte F, Erqou S, Laukkanen J, et al. Meta-analysis of ventricular premature complexes and their relation to cardiac
mortality in general populations. Am J Cardiol. 2013;112:1263-70.
10.Lin CY, Chang SL, Lin YJ, et al. Long-term outcome of multiform premature ventricular complexes in structurally
normal heart. Int J Cardiol. 2015;180:80-5.
11.Lin CY, Chang SL, Chung FP, et al. Long-term outcome of non-sustained ventricular tachycardia in structurally normal
hearts. PloS One. 2016;11:e0160181.
12.Ruberman W, Weinblatt E, Goldberg JD, et al. Ventricular premature complexes and sudden death after myocardial
infarction. Circulation. 1981;64:297-305.
13.Ruberman W, Weinblatt E, Goldberg JD, et al. Ventricular premature beats and mortality after myocardial infarction.
N Engl J Med. 1977;297:750-7.
14.The Cardiac Arrhythmia Suppression Trial II Investigators. Effect of the antiarrhythmic agent moricizine on survival
after myocardial infarction. The Cardiac Arrhythmia Suppression Trial II Investigators. N Engl J Med. 1992;327:227-
33.
15.Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in patients receiving encainide, flecainide, or
placebo. The Cardiac Arrhythmia Suppression Trial. N Engl J Med. 1991;324:781-8.
16.Morganroth J Goin JE. Quinidine-related mortality in the short-to-medium-term treatment of ventricular
arrhythmias. A meta-analysis. Circulation. 1991;84:1977-83.

Page 21
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

17.Waldo AL, Camm AJ, deRuyter H, et al. Effect of d-sotalol on mortality in patients with left ventricular dysfunction
after recent and remote myocardial infarction. The SWORD Investigators. Survival With Oral d-Sotalol. Lancet.
1996;348:7-12.
18.Ling Z, Liu Z, Su L, et al. Radiofrequency ablation versus antiarrhythmic medication for treatment of ventricular
premature beats from the right ventricular outflow tract: prospective randomized study. Circ Arrhythm
Electrophysiol. 2014;7:237-43.
19.Jouven X, Zureik M, Desnos M, et al. Long-term outcome in asymptomatic men with exercise-induced premature
ventricular depolarizations. N Engl J Med. 2000;343:826-33.
20.Frolkis JP, Pothier CE, Blackstone EH, et al. Frequent ventricular ectopy after exercise as a predictor of death. N Engl
J Med. 2003;348:781-90.
21.Biffi A, Pelliccia A, Verdile L, et al. Long-term clinical significance of frequent and complex ventricular
tachyarrhythmias in trained athletes. J Am Coll Cardiol. 2002;40:446-52.
22.Heidbchel H, Hoogsteen J, Fagard R, et al. High prevalence of right ventricular involvementin endurance athletes
with ventricular arrhythmiasRole of an electrophysiologic study in risk stratification. Eur Heart J. 2003;24:1473-80.
23.Kanei Y, Friedman M, Ogawa N, et al. Frequent premature ventricular complexes originating from the right
ventricular outflow tract are associated with left ventricular dysfunction. Ann Noninvasive Electrocardiol.
2008;13:81-5.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

24.Lee GK, Klarich KW, Grogan M, et al. Premature ventricular contraction-induced cardiomyopathy: a treatable
condition. Circ Arrhythm Electrophysiol. 2012;5:229-36.
25.Viskin S, Rosso R, Rogowski O, et al. The "short-coupled" variant of right ventricular outflow ventricular tachycardia:
a not-so-benign form of benign ventricular tachycardia? J Cardiovasc Electrophysiol. 2005;16:912-6.
26.Noda T, Shimizu W, Taguchi A, et al. Malignant entity of idiopathic ventricular fibrillation and polymorphic
ventricular tachycardia initiated by premature extrasystoles originating from the right ventricular outflow tract. J
Am Coll Cardiol. 2005;46:1288-94.
27.Dumas F, Cariou A, Manzo-Silberman S, et al. Immediate percutaneous coronary intervention is associated with
better survival after out-of-hospital cardiac arrest: insights from the PROCAT (Parisian Region Out of hospital Cardiac
ArresT) registry. Circ Cardiovasc Interv. 2010;3:200-7.
28.Gupta S, Pressman GS, Figueredo VM. Incidence of, predictors for, and mortality associated with malignant
ventricular arrhythmias in non-ST elevation myocardial infarction patients. Coron. Artery Dis. 2010;21:460-5.
29.Terkelsen CJ, Sorensen JT, Kaltoft AK, et al. Prevalence and significance of accelerated idioventricular rhythm in
patients with ST-elevation myocardial infarction treated with primary percutaneous coronary intervention. Am J
Cardiol. 2009;104:1641-6.
30.Al-Khatib SM, Granger CB, Huang Y, et al. Sustained ventricular arrhythmias among patients with acute coronary
syndromes with no ST-segment elevation: incidence, predictors, and outcomes. Circulation. 2002;106:309-12.
31.Jabbari R, Engstrom T, Glinge C, et al. Incidence and risk factors of ventricular fibrillation before primary angioplasty
in patients with first ST-elevation myocardial infarction: a nationwide study in Denmark. J Am Heart Assoc.
2015;4:e001399.
32.Mehta RH, Starr AZ, Lopes RD, et al. Incidence of and outcomes associated with ventricular tachycardia or fibrillation
in patients undergoing primary percutaneous coronary intervention. JAMA. 2009;301:1779-89.
33.Volpi A, Cavalli A, Franzosi MG, et al. One-year prognosis of primary ventricular fibrillation complicating acute
myocardial infarction. The GISSI (Gruppo Italiano per lo Studio della Streptochinasi nell'Infarto miocardico)
investigators. Am J Cardiol. 1989;63:1174-8.
34.El-Sherif N, Smith RA, Evans K. Canine ventricular arrhythmias in the late myocardial infarction period. 8. Epicardial
mapping of reentrant circuits. Circ Res. 1981;49:255-65.
35.Ackerman MJ, Priori SG, Willems S, et al. HRS/EHRA expert consensus statement on the state of genetic testing for
the channelopathies and cardiomyopathies: this document was developed as a partnership between the Heart
Rhythm Society (HRS) and the European Heart Rhythm Association (EHRA). Europace. 2011;13:1077-109.
36.Nannenberg EA, Sijbrands EJ, Dijksman LM, et al. Mortality of inherited arrhythmia syndromes: insight into their
natural history. Circ Cardiovasc Genet. 2012;5:183-9.
37.Hassaguerre M, Shah DC, Jais P, et al. Role of Purkinje conducting system in triggering of idiopathic ventricular
fibrillation. Lancet. 2002;359:677-8.
38.Hassaguerre M, Shoda M, Jais P, et al. Mapping and ablation of idiopathic ventricular fibrillation. Circulation.
2002;106:962-7.

Page 22
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

39.Goldberger JJ, Buxton AE, Cain M, et al. Risk stratification for arrhythmic sudden cardiac death: identifying the
roadblocks. Circulation. 2011;123:2423-30.
40.Fishman GI, Chugh SS, DiMarco JP, et al. Sudden cardiac death prediction and prevention: report from a National
Heart, Lung, and Blood Institute and Heart Rhythm Society Workshop. Circulation. 2010;122:2335-48.
41.Myerburg RJ. Sudden cardiac death: exploring the limits of our knowledge. J Cardiovasc Electrophysiol. 2001;12:369-
81.
42.Kong MH, Fonarow GC, Peterson ED, et al. Systematic review of the incidence of sudden cardiac death in the United
States. J Am Coll Cardiol. 2011;57:794-801.
43.Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke statistics2016 update: a report from the
American Heart Association. Circulation. 2016;133:e38-360.
44.Benjamin EJ, Blaha MJ, Chiuve SE, et al. Heart disease and stroke statistics2017 update: a report from the
American Heart Association. Circulation. 2017;135:e146-603.
45.Merchant RM, Yang L, Becker LB, et al. Incidence of treated cardiac arrest in hospitalized patients in the United
States. Crit Care Med. 2011;39:2401-6.
46. Institute of Medicine. Committee on the Treatment of Cardiac Arrest: current status and future directions:
strategies to improve cardiac arrest survival: a time to act. Washington, DC: National Academic Press; 2015.
47.Jollis JG, Granger CB. Improving care of out-of-hospital cardiac arrest: next steps. Circulation. 2016;134:2040-2.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

48.Daya MR, Schmicker R, May SH, et al. Current burden of cardiac arrest in the United States: report from the
Resuscitation Outcomes Consortium. Paper commissioned by the Committee on the Treatment of Cardiac Arrest:
Current Status and Future Directions. 2015.
49.Myerburg RJ, Halperin H, Egan DA, et al. Pulseless electric activity: definition, causes, mechanisms, management,
and research priorities for the next decade: report from a National Heart, Lung, and Blood Institute workshop.
Circulation. 2013;128:2532-41.
50.Myerburg RJ, Goldberger JJ. Sudden cardiac arrest risk assessment: Population science and the individual risk
mandate. JAMA Cardiol. 2017;2:689-94.
51.Bogle BM, Ning H, Mehrotra S, et al. Lifetime risk for sudden cardiac death in the community. J Am Heart Assoc.
2016;5:e002398.
52.Junttila MJ, Hookana E, Kaikkonen KS, et al. Temporal trends in the clinical and pathological characteristics of victims
of sudden cardiac death in the absence of previously identified heart disease. Circ Arrhythm Electrophysiol.
2016;9:e003723.
53.Hookana E, Junttila MJ, Puurunen VP, et al. Causes of nonischemic sudden cardiac death in the current era. Heart
Rhythm. 2011;8:1570-5.
54.Wong MK, Morrison LJ, Qiu F, et al. Trends in short- and long-term survival among out-of-hospital cardiac arrest
patients alive at hospital arrival. Circulation. 2014;130:1883-90.

3. General Evaluation of Patients With Documented or Suspected VA


3.1. History and Physical Examination
Recommendation for Syncope*
Referenced studies that support the recommendation are summarized in Online Data Supplement 1.
COR LOE Recommendation
1. Patients presenting with syncope for which VA is documented, or thought to
I B-NR be a likely cause, should be hospitalized for evaluation, monitoring, and
management (1-4).
*This section covers practices that are well accepted, and a new recommendation was determined to only be warranted
for syncope.

Table 6

Page 23
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 6. Important Considerations in the Evaluation of Patients With Known or Suspected VA

Component Assessment and Findings Relevant for VA and/or SCD Risk


History 1. Symptoms/events related to arrhythmia: Palpitations, lightheadedness, syncope, dyspnea,
chest pain, cardiac arrest
2. Symptoms related to underlying heart disease: Dyspnea at rest or on exertion, orthopnea,
paroxysmal nocturnal dyspnea, chest pain, edema
3. Precipitating factors: Exercise, emotional stress
4. Known heart disease: Coronary, valvular (e.g., mitral valve prolapse), congenital heart
disease, other
5. Risk factors for heart disease: Hypertension, diabetes mellitus, hyperlipidemia, and smoking
6. Medications:
Antiarrhythmic medications
Other medications with potential for QT prolongation and torsades de pointes
Medications with potential to provoke or aggravate VA
Stimulants including cocaine and amphetamines
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Supplements including anabolic steroids


Medication-medication interaction that could cause QT prolongation and torsades de
pointes
7. Past medical history:
Thyroid disease
Acute kidney injury, chronic kidney disease, or electrolyte abnormalities
Stroke or embolic events
Lung disease
Epilepsy (arrhythmic syncope can be misdiagnosed as epilepsy)
Alcohol or illicit drug use
Use of over-the-counter medications that could cause QT prolongation and torsades de
pointes
Unexplained motor vehicle crashes
Family History 1. SCD, SCA, or unexplained drowning in a first-degree relative
2. SIDS or repetitive spontaneous pregnancy losses given their potential association with
cardiac channelopathies
3. Heart disease
IHD
Cardiomyopathy: Hypertrophic, dilated, ARVC
Congenital heart disease
Cardiac channelopathies: Long QT, Brugada, Short QT, CPVT
Arrhythmias
Conduction disorders, pacemakers/ICDs
4. Neuromuscular disease associated with cardiomyopathies
Muscular dystrophy
5. Epilepsy
Examination 1. Heart rate and regularity, blood pressure
2. Jugular venous pressure
3. Murmurs
4. Pulses and bruits
5. Edema
6. Sternotomy scars
ARVC indicates arrhythmogenic right ventricular cardiomyopathy; CPVT catecholaminergic polymorphic ventricular
tachycardia; IHD, ischemic heart disease; SCA, sudden cardiac arrest; SCD, sudden cardiac death; SIDS, sudden infant
death syndrome; and VA, ventricular arrhythmia.

Page 24
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

References
1. Middlekauff HR, Stevenson WG, Stevenson LW, et al. Syncope in advanced heart failure: high risk of sudden death
regardless of origin of syncope. J Am Coll Cardiol. 1993;21:110-6.
2. Myerburg RJ, Castellanos A. Cardiac arrest and sudden cardiac death - Chapter 39. In: Mann DL, Zipes DP, Libby P, et
al, eds. Braunwald's Heart Disease: A Textbook of Cardiovascular Medicine. 10th ed. Oxford, UK: Elsevier; 2015:821-
60.
3. Shen WK, Sheldon RS, Benditt DG, et al. 2017 ACC/AHA/HRS guideline for the evaluation and management of
patients with syncope: a report of the American College of Cardiology/American Heart Association Task Force on
Clinical Practice Guidelines and the Heart Rhythm Society. Circulation. 2017;136:e60-122.
4. Soteriades ES, Evans JC, Larson MG, et al. Incidence and prognosis of syncope. N Engl J Med. 2002;347:878-85.

3.2. Noninvasive Evaluation


3.2.1. 12-lead ECG and Exercise Testing
Recommendations for 12-lead ECG and Exercise Testing
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

References studies that support the recommendations are summarized in Online Data Supplement 2.
COR LOE Recommendations
1. In patients with sustained, hemodynamically stable, wide complex
I B-NR
tachycardia, a 12-lead ECG during tachycardia should be obtained (1-3).
2. In patients with VA symptoms associated with exertion, suspected ischemic
I B-NR heart disease, or catecholaminergic polymorphic ventricular tachycardia,
exercise treadmill testing is useful to assess for exercise-induced VA (4, 5).
3. In patients with suspected or documented VA, a 12-lead ECG should be
I B-NR
obtained in sinus rhythm to look for evidence of heart disease (6).

References
1. Brugada P, Brugada J, Mont L, et al. A new approach to the differential diagnosis of a regular tachycardia with a
wide QRS complex. Circulation. 1991;83:1649-59.
2. Wellens HJ, Bar FW, Lie KI. The value of the electrocardiogram in the differential diagnosis of a tachycardia with a
widened QRS complex. Am J Med. 1978;64:27-33.
3. Steinman RT, Herrera C, Schuger CD, et al. Wide QRS tachycardia in the conscious adult. Ventricular tachycardia is
the most frequent cause. JAMA. 1989;261:1013-6.
4. Elhendy A, Chandrasekaran K, Gersh BJ, et al. Functional and prognostic significance of exercise-induced ventricular
arrhythmias in patients with suspected coronary artery disease. Am J Cardiol. 2002;90:95-100.
5. Grady TA, Chiu AC, Snader CE, et al. Prognostic significance of exercise-induced left bundle-branch block. JAMA.
1998;279:153-6.
6. Perez-Rodon J, Martinez-Alday J, Baron-Esquivias G, et al. Prognostic value of the electrocardiogram in patients with
syncope: data from the group for syncope study in the emergency room (GESINUR). Heart Rhythm. 2014;11:2035-
44.

Page 25
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

3.2.2. Ambulatory Electrocardiography


Recommendation for Ambulatory Electrocardiography
Referenced studies that support the recommendation are summarized in Online Data Supplement 3 and
4.
COR LOE Recommendation
1. Ambulatory electrocardiographic monitoring is useful to evaluate whether
I B-NR symptoms, including palpitations, presyncope, or syncope, are caused by VA
(1-4).

References
1. Barrett PM, Komatireddy R, Haaser S, et al. Comparison of 24-hour Holter monitoring with 14-day novel adhesive
patch electrocardiographic monitoring. Am J Med. 2014;127:95-7.
2. de Asmundis C, Conte G, Sieira J, et al. Comparison of the patient-activated event recording system vs. traditional 24
h Holter electrocardiography in individuals with paroxysmal palpitations or dizziness. Europace. 2014;16:1231-5.
3. Linzer M, Pritchett EL, Pontinen M, et al. Incremental diagnostic yield of loop electrocardiographic recorders in
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

unexplained syncope. Am J Cardiol. 1990;66:214-9.


4. Turakhia MP, Hoang DD, Zimetbaum P, et al. Diagnostic utility of a novel leadless arrhythmia monitoring device. Am
J Cardiol. 2013;112:520-4.

3.2.3. Implanted Cardiac Monitors


Recommendation for Implanted Cardiac Monitors
Referenced studies that support the recommendation are summarized in Online Data Supplement 5.
COR LOE Recommendation
1. In patients with sporadic symptoms (including syncope) suspected to be
IIa B-R
related to VA, implanted cardiac monitors can be useful (1-4).

References
1. Bloch Thomsen PE, Jons C, Raatikainen MJ, et al. Long-term recording of cardiac arrhythmias with an implantable
cardiac monitor in patients with reduced ejection fraction after acute myocardial infarction: the Cardiac
Arrhythmias and Risk Stratification After Acute Myocardial Infarction (CARISMA) study. Circulation. 2010;122:1258-
64.
2. Krahn AD, Klein GJ, Yee R, et al. Use of an extended monitoring strategy in patients with problematic syncope.
Reveal Investigators. Circulation. 1999;99:406-10.
3. Solbiati M, Costantino G, Casazza G, et al. Implantable loop recorder versus conventional diagnostic workup for
unexplained recurrent syncope. Cochrane Database Syst Rev. 2016;4:CD011637.
4. Volosin K, Stadler RW, Wyszynski R, et al. Tachycardia detection performance of implantable loop recorders: results
from a large 'real-life' patient cohort and patients with induced ventricular arrhythmias. Europace. 2013;15:1215-22.

Page 26
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

3.2.4. Noninvasive Cardiac Imaging


Recommendations for Noninvasive Cardiac Imaging
Referenced studies that support the recommendations are summarized in Online Data Supplement 6.
COR LOE Recommendations
1. In patients with known or suspected VA that may be associated with
I B-NR underlying structural heart disease or a risk of SCA, echocardiography is
recommended for evaluation of cardiac structure and function (1, 2).
2. In patients presenting with VA who are suspected of having structural heart
disease, cardiac magnetic resonance imaging (MRI) or computed tomography
IIa C-EO
(CT) can be useful to detect and characterize underlying structural heart
disease.

References
1. Solomon SD, Zelenkofske S, McMurray JJ, et al. Sudden death in patients with myocardial infarction and left
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

ventricular dysfunction, heart failure, or both. N Engl J Med. 2005;352:2581-8.


2. Gula LJ, Klein GJ, Hellkamp AS, et al. Ejection fraction assessment and survival: an analysis of the Sudden Cardiac
Death in Heart Failure Trial (SCD-HeFT). Am Heart J. 2008;156:1196-200.

3.2.5. Biomarkers
Recommendation for Biomarkers
Referenced studies that support the recommendation are summarized in Online Data Supplement 7.
COR LOE Recommendation
1. In patients with structural heart disease, measurement of natriuretic peptides
IIa B-NR (BNP or N-terminal pro-BNP) can be useful by adding prognostic information
to standard risk factors for predicting SCD or SCA (1-4).

References
1. Ahmad T, Fiuzat M, Neely B, et al. Biomarkers of myocardial stress and fibrosis as predictors of mode of death in
patients with chronic heart failure. JACC Heart Fail. 2014;2:260-8.
2. Scott PA, Barry J, Roberts PR, et al. Brain natriuretic peptide for the prediction of sudden cardiac death and
ventricular arrhythmias: a meta-analysis. Eur J Heart Fail. 2009;11:958-66.
3. Levine YC, Rosenberg MA, Mittleman M, et al. B-type natriuretic peptide is a major predictor of ventricular
tachyarrhythmias. Heart Rhythm. 2014;11:1109-16.
4. Berger R, Huelsman M, Strecker K, et al. B-type natriuretic peptide predicts sudden death in patients with chronic
heart failure. Circulation. 2002;105:2392-7.

3.2.6. Genetic Considerations in Arrhythmia Syndromes

Recommendation for Genetic Counselling*


COR LOE Recommendation
1. In patients and family members in whom genetic testing for risk stratification
I C-EO
for SCA or SCD is recommended, genetic counseling is beneficial.
*Please refer to section 7.9 in the full guideline for disease-specific recommendations.

Page 27
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

3.3. Invasive Testing


3.3.1. Invasive Cardiac Imaging: Cardiac Catheterization or CT Angiography
Recommendation for Invasive Imaging: Cardiac Catheterization
COR LOE Recommendation
1. In patients who have recovered from unexplained SCA, CT or invasive coronary
I C-EO angiography is useful to confirm the presence or absence of ischemic heart
disease and guide decisions for myocardial revascularization.

3.3.2. Electrophysiological Study for VA


Recommendations for Electrophysiological Study
References that support the recommendations are summarized in Online Data Supplement 8 and 9.
COR LOE Recommendations
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

1. In patients with ischemic cardiomyopathy, NICM, or adult congenital heart


disease who have syncope or other VA symptoms and who do not meet
IIa B-R
indications for a primary prevention ICD, an electrophysiological study can
be useful for assessing the risk of sustained VT (1-7).
2. In patients who meet criteria for ICD implantation, an electrophysiological
III: No
B-R study for the sole reason of inducing VA is not indicated for risk stratification
Benefit
(8-11).
3. An electrophysiological study is not recommended for risk stratification for
III: No VA in the setting of long QT syndrome, catecholaminergic polymorphic
B-NR
Benefit ventricular tachycardia, short QT syndrome, or early repolarization
syndromes (12-16).

References
1. Buxton AE, Lee KL, DiCarlo L, et al. Electrophysiologic testing to identify patients with coronary artery disease who
are at risk for sudden death. Multicenter Unsustained Tachycardia Trial Investigators. N Engl J Med 2000;342:1937-
45.
2. Buxton AE, Lee KL, Hafley GE, et al. Relation of ejection fraction and inducible ventricular tachycardia to mode of
death in patients with coronary artery disease: an analysis of patients enrolled in the multicenter unsustained
tachycardia trial. Circulation. 2002;106:2466-72.
3. Costantini O, Hohnloser SH, Kirk MM, et al. The ABCD (Alternans Before Cardioverter Defibrillator) Trial: strategies
using T-wave alternans to improve efficiency of sudden cardiac death prevention. J Am Coll Cardiol. 2009;53:471-9.
4. Bourke JP, Richards DA, Ross DL, et al. Routine programmed electrical stimulation in survivors of acute myocardial
infarction for prediction of spontaneous ventricular tachyarrhythmias during follow-up: results, optimal stimulation
protocol and cost-effective screening. J Am Coll Cardiol. 1991;18:780-8.
5. Bailey JJ, Berson AS, Handelsman H, et al. Utility of current risk stratification tests for predicting major arrhythmic
events after myocardial infarction. J Am Coll Cardiol. 2001;38:1902-11.
6. Schmitt C, Barthel P, Ndrepepa G, et al. Value of programmed ventricular stimulation for prophylactic internal
cardioverter-defibrillator implantation in postinfarction patients preselected by noninvasive risk stratifiers. J Am Coll
Cardiol. 2001;37:1901-7.
7. Hilfiker G, Schoenenberger AW, Erne P, et al. Utility of electrophysiological studies to predict arrhythmic events.
World J Cardiol. 2015;7:344-50.
8. Bardy GH, Lee KL, Mark DB, et al. Amiodarone or an implantable cardioverter-defibrillator for congestive heart
failure. N Engl J Med. 2005;352:225-37.

Page 28
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

9. Buxton AE, Lee KL, Fisher JD, et al. A randomized study of the prevention of sudden death in patients with coronary
artery disease. Multicenter Unsustained Tachycardia Trial Investigators. N Engl J Med. 1999;341:1882-90.
10.Moss AJ, Hall WJ, Cannom DS, et al. Improved survival with an implanted defibrillator in patients with coronary
disease at high risk for ventricular arrhythmia. Multicenter Automatic Defibrillator Implantation Trial Investigators.
N Engl J Med. 1996;335:1933-40.
11.Moss AJ, Zareba W, Hall WJ, et al. Prophylactic implantation of a defibrillator in patients with myocardial infarction
and reduced ejection fraction. N Engl J Med. 2002;346:877-83.
12.Bhandari AK, Shapiro WA, Morady F, et al. Electrophysiologic testing in patients with the long QT syndrome.
Circulation. 1985;71:63-71.
13.Giustetto C, Di MF, Wolpert C, et al. Short QT syndrome: clinical findings and diagnostic-therapeutic implications.
Eur. Heart J. 2006;27:2440-7.
14.Giustetto C, Schimpf R, Mazzanti A, et al. Long-term follow-up of patients with short QT syndrome. J Am Coll Cardiol.
2011;58:587-95.
15.Mahida S, Derval N, Sacher F, et al. Role of electrophysiological studies in predicting risk of ventricular arrhythmia in
early repolarization syndrome. J Am Coll Cardiol. 2015;65:151-9.
16.Priori SG, Napolitano C, Memmi M, et al. Clinical and molecular characterization of patients with catecholaminergic
polymorphic ventricular tachycardia. Circulation. 2002;106:69-74.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

4. Therapies for Treatment or Prevention of VA


4.1. Medication Therapy
Table 7
Table 7. Pharmacological Characteristics of Available Antiarrhythmic Medications for Treating VA
Antiarrhythmic
Medication
(Class) and Uses in Electrophysiological Pharmacological Common Adverse
Dose VA/SCA Target Effects Characteristics Effects
Acebutolol VT, PVCs Beta 1, Sinus rate slowed Active Cardiac: Bradycardia,
PO 2001200 Mild intrinsic AV nodal metabolite t1/2: hypotension, HF,
mg daily or sympathomimetic refractoriness 813 h AVB
upto 600 mg activity increased pProlonged with Other: Dizziness,
bid renal fatigue, anxiety,
impairment) impotence,
Metab: H hyper/hypoesthesia
Excr: F 60%, U
40%
Amiodarone VT, VF, I Na, I Ca, I Kr, I K1, I Ks, Sinus rate slowed t1/2: 26-107 d Cardiac:
(III) PVC, I to, Beta receptor, QRS prolonged Metab: H Hypotension,
Alpha receptor QTc prolonged Excr: F bradycardia, AVB,
IV: 300 mg nuclear T3 AV nodal TdP, slowsVT below
bolus for VF/ receptor refractoriness programmed ICD
pulseless VT increased; detection rate,
arrest; 150-mg increased DFT increases
bolus for stable defibrillation
VT; 1 mg/min x threshold
6 h, then 0.5
mg/min x 18 h Other: Corneal
microdeposits,
PO: 400 mg* q thyroid
8 to 12 h for 1 abnormalities,
2 wk, then 300- ataxia, nausea,

Page 29
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

400 mg daily; emesis, constipation,


reduce dose to photosensitivity, skin
200 mg daily if discoloration, ataxia,
possible dizziness, peripheral
neuropathy, tremor,
hepatitis, cirrhosis,
pulmonary fibrosis
or pneumonitis
Atenolol (II) VT, PVC, Beta 1 Sinus rate slowed t1/2: 67 h Cardiac: Bradycardia,
ARVC, AV nodal (prolonged with hypotension, HF,
PO: 25100 mg LQTS refractoriness renal AVB
qd or bid increased impairment) Other: Dizziness,
Metab: H fatigue, depression,
Excr: F 50%, U impotence
40%
Bisoprolol (II) VT, PVC Beta 1 receptor Sinus rate slowed t1/2: 912 h Cardiac: Chest pain,
AV nodal Metab: H bradycardia, AVB
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

PO: 2.510 mg refractoriness Excr: U Other: Fatigue,


once daily increased insomnia, diarrhea
Carvedilol (II) VT, PVC Beta 1 and 2 Sinus rate slowed t1/2: 710 h Cardiac: Bradycardia,
receptors, Alpha AV nodal Metab: H hypotension, AVB,
PO: 3.12525 refractoriness Excr: F edema, syncope
mg q 12 h increased Other:
Hyperglycemia,
dizziness, fatigue,
diarrhea
Diltiazem (IV) VT I Ca-L Sinus rate slowed t1/2: Injection 2 Cardiac:
specifically PR prolonged 5 h, immediate Hypotension,
IV: 5-10 mg RVOT, AV nodal release 4.512 edema, HF, AVB,
Qq 15-30 min idiopathic conduction slowed h, extended bradycardia,
LVT release 12 h, exacerbation of
Extended and severe HFrEF
release: PO: hepatic Other: Headache,
120360 impairment 14 rash, constipation
mg/day 16 h
Metab: H
Excr: U
Esmolol (II) VT Beta 1 receptor Sinus rate slowed t1/2: 9 min Cardiac: Bradycardia,
AV nodal Metab: RBC hypotension, HF,
IV: 0.5 mg/kg refractoriness esterases AVB
bolus, 0.05 increased Excr: U Other: Dizziness,
mg/kg/min nausea
Flecainide (IC)
VT, PVC (in I Na, I Kr, I Kur PR prolonged t1/2: 722 h Cardiac: Sinus node
the QRS prolonged; Metab: H dysfunction, AVB,
PO: 50200 mg absence of increased DFT Excr: U drug-induced
q 12 h structural Brugada syndrome.
heart monomorphic VT in
disease). patients with a
Has a role myocardial scar,
in treating exacerbation of
patients HFrEF
with CPVT Other: Dizziness,
tremor, vision

Page 30
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

disturbance,
dyspnea, nausea
Lidocaine (IB) VT, VF I Na No marked effect Initial t1/2 730 Cardiac :
on most intervals; min; terminal Bradycardia,
IV: 1 mg/kg QTc can slightly 90120 min. hemodynamic
bolus, 13 shorten Prolonged in HF, collapse, AVB, sinus
mg/min liver disease, arrest
shock, severe Other: Delirium,
1-1.5 mg/kg. renal disease psychosis, seizure,
Repeat 0.5 Metab: H nausea, tinnitus,
0.75 mg/kg Excr: U dyspnea,
bolus every 5 bronchospasm
10 min (max
cumulative
dose 3 mg/kg).
Maintenance
infusion is 14
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

mg/min
although one
could start at
0.5 mg/min
Metoprolol (II) VT, PVC Beta 1 receptor Sinus rate slowed t1/2: 34 h Cardiac: Bradycardia,
AV nodal Metab: H hypotension, AVB
IV: 5 mg q 5 refractoriness Excr: U Other: Dizziness,
min up to 3 increased fatigue, diarrhea,
doses depression, dyspnea

PO: 25100 mg
Extended
release qd or q
12 h
Mexiletine (IB) T, VF, PVC, I Na No marked effect t1/2: 1014 h Cardiac: HF, AVB
has a role on most intervals; Metab: H Other: Lightheaded,
PO: 150300 in patients QTc can slightly Excr: U tremor, ataxia,
mg q 8 h or q with LQT3 shorten paresthesias,
12 h nausea, blood
dyscrasias
Nadolol (II) VT, PVC, Beta 1 and 2 Sinus rate slowed t1/2: 2024 h Cardiac: Bradycardia,
LQTS, CPVT receptors AV nodal Metab: none hypotension, HF,
PO: 40320 mg refractoriness Excr: U AVB
daily increased Other: Edema,
dizziness, cold
extremities,
bronchospasm
Procainamide VT I Na, I Kr QRS prolonged Metab: H Cardiac: TdP; AVB,
(IA) QTc prolonged; t1/2: 25 h; NAPA hypotension and
increased DFT 68 h exacerbation of
IV: loading t1/2 prolonged in HFrEF
dose 1017 renal Other: Lupus
mg/kg at 20 dysfunction. symptoms, diarrhea,
50 mg/min Anephric: proc nausea, blood
11 h and NAPA dyscrasias
42 h

Page 31
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Maintenance Excr: U
dose: 14
mg/min
PO (SR
preparation):
5001250 mg q
6h
Propafenone VT, PVC (in I Na, I Kr, I Kur, Beta PR prolonged t1/2: 210 h or Cardiac: HF, AVB,
(IC) the receptor, Alpha QRS prolonged; 1032 h drug-induced
absence of receptor increased DFT t1/2: extensive Brugada syndrome
PO: Immediate structural metabolizers 2 Other: Dizziness,
release 150 heart 10 h; poor fatigue, nausea,
300 mg q 8 h disease) metabolizers diarrhea,
Extended 1032 h. xerostomia, tremor,
release 225 Metab: H blurred vision
425 mg q 12 h Excr: U
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Propranolol (II) VT, PVC, Beta 1 and 2 Sinus rate slowed t1/2: Immediate Cardiac: Bradycardia,
LQTS receptors, I Na AV nodal release 36 h hypotension, HF,
IV: 13 mg q 5 refractoriness Extended AVB
min to a total increased release 810 h Other: Sleep
of 5 mg Metab: H disorder, dizziness,
Excr: U nightmares,
PO: Immediate hyperglycemia,
release 1040 diarrhea,
mg q 6 h; bronchospasm
Extended
release 60160
mg q 12 h
Quinidine (IA) T, VF, I Na, I to, I Kr, M, QRS prolonged : 6-8 h longer
1/2 Cardiac: Syncope,
(including Alpha receptor QTc prolonged; in HF, liver TdP, AVB
PO: sulfate salt short QT increased DFT cirrhosis, and Other: Dizziness,
200600 mg q syndrome, with older age diarrhea, nausea,
6 h to q 12 h Brugada) Metab: H esophagitis, emesis,
Excr: U tinnitus, blurred
gGluconate salt vision, rash,
324648 mg q weakness, tremor;
8 h to q 12 h blood dyscrasias

IV: loading
dose: 800 mg
in 50 mL
infused at 50
mg/min
Ranolazine VT I Na, I Kr Sinus rate slowed t1/2: 7 h Cardiac: Bradycardia,
(not classified) Tc prolonged Metab: H hypotension
Excr: U 75%, F Other: Headache,
PO: 5001000 25% dizziness, syncope,
mg q 12 h nausea, dyspnea
Sotalol (III) VT, VF, PVC I Kr, Beta 1 and 2 Sinus rate slowed t1/2: 12 h Cardiac: Bradycardia,
receptor QTc prolonged Metab: none hypotension, HF,
IV: 75 mg q 12 AV nodal Excr: U syncope, TdP
h refractoriness

Page 32
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

increased; Other: Fatigue,


PO: 80120 mg decreased DFT dizziness, weakness,
q 12 h, may dyspnea, bronchitis,
increase dose depression, nausea,
every 3 d; max diarrhea
320 mg/d
Verapamil (IV) VT I Ca-L Sinus rate slowed t1/2: 37 h Cardiac:
(specifically PR prolonged Metab: H Hypotension,
IV: 2.55 mg q RVOT, AV nodal Excr: U edema, HF, AVB,
1530 min verapamil- conduction slowed bradycardia,
sensitive exacerbation of
Sustained idiopathic HFrEF
release PO: LVT) Other: Headache,
240480 mg/d rash, gingival
hyperplasia,
constipation,
dyspepsia
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

*Although up to 800 mg every 8 h might be used, higher doses of amiodarone are associated with a higher risk of adverse
events.
Alpha indicates alpha-adrenergic receptor; ARVC, arrhythmogenic right ventricular cardiomyopathy; AV, atrioventricular;
AVB, atrioventricular block; Beta, beta-adrenergic receptor; HF, heart failure; CPVT, catecholaminergic polymorphic
ventricular tachycardia; DFT, defibrillation threshold; F, feces; H, hepatic; I Ca, L-type calcium channel current; I K1, inward
rectifier potassium channel; I KACh, muscarinic receptor-gated potassium channel; I KATP, adenosine-activated potassium
channel; I Kr, rapid delayed rectifier potassium current; I Ks, slow delayed rectifier potassium current; I Kur, ultra-rapid
delayed rectifier potassium current; I Na, fast inward sodium current; I to, transient outward potassium current; LQTS, long-
QT syndrome; LVT, left ventricular tachycardia; M, muscarinic; Metab, metabolism; NAPA, n-acetyl procainamide; PVC,
premature ventricular complex; QTc, corrected QT interval; t1/2, half-life; RVOT, right ventricular outflow tract; T3,
triiodothyronine; TdP, torsades de pointes; U, urine; VT, ventricular tachycardia; and VF, ventricular fibrillation.
Modified from Shleifer JW, et al (1).

Reference
1. Schleifer JW, Sorajja D, Shen WK. Advances in the pharmacologic treatment of ventricular arrhythmias. Expert Opin
Pharmacother. 2015;16:2637-51.

4.2. Preventing SCD With HF Medications


Recommendation for Pharmacological Prevention of SCD
References that support the recommendation are summarized in Online Data Supplement 10.
COR LOE Recommendation
1. In patients with HFrEF (LVEF 40%), treatment with a beta blocker, a
mineralocorticoid receptor antagonist and either an angiotensin-converting
I A enzyme inhibitor, an angiotensin-receptor blocker, or an angiotensin receptor-
neprilysin inhibitor is recommended to reduce SCD and all-cause mortality (1-
8).

References
1. Yancy CW, Jessup M, Bozkurt B, et al. 2016 ACC/AHA/HFSA Focused update on new pharmacological therapy for
heart failure: an update of the 2013 ACCF/AHA guideline for the management of heart failure: a report of the
American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart
Failure Society of America. Circulation. 2016;134:e282-93.
2. Committees for Cardiac insufficiency Bisoprolol Study II. The cardiac insufficiency Bisoprolol Study II (CIBIS-II): a
randomised trial. Lancet. 1999;353:9-13.

Page 33
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

3. Cohn JN, Johnson G, Ziesche S, et al. A comparison of enalapril with hydralazine-isosorbide dinitrate in the
treatment of chronic congestive heart failure. N Engl J Med. 1991;325:303-10.
4. Packer M, Bristow MR, Cohn JN, et al. The effect of carvedilol on morbidity and mortality in patients with chronic
heart failure. U.S. Carvedilol Heart Failure Study Group. N Engl J Med. 1996;334:1349-55.
5. Dargie HJ. Effect of carvedilol on outcome after myocardial infarction in patients with left-ventricular dysfunction:
the CAPRICORN randomised trial. Lancet. 2001;357:1385-90.
6. Pitt B, Remme W, Zannad F, et al. Eplerenone, a selective aldosterone blocker, in patients with left ventricular
dysfunction after myocardial infarction. N Engl J Med. 2003;348:1309-21.
7. Al Chekakie MO. Traditional heart failure medications and sudden cardiac death prevention: a review. J Cardiovasc
Pharmacol Ther. 2013;18:412-26.
8. Pfeffer MA, McMurray JJ, Velazquez EJ, et al. Valsartan, captopril, or both in myocardial infarction complicated by
heart failure, left ventricular dysfunction, or both. N Engl J Med. 2003;349:1893-906.

4.3. Surgery and Revascularization Procedures in Patients With Ischemic Heart


Disease
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Recommendations for Surgery and Revascularization Procedures in Patients With Ischemic


Heart Disease
References that support the recommendations are summarized in Online Data Supplement 11.
COR LOE Recommendations
1. Patients with sustained VA and survivors of SCA should be evaluated for
I B-NR
ischemic heart disease, and should be revascularized as appropriate (1-4).
2. In patients with anomalous origin of a coronary artery suspected to be the
I C-EO
cause of SCA, repair or revascularization is recommended.

References
1. Cook JR, Rizo-Patron C, Curtis AB, et al. Effect of surgical revascularization in patients with coronary artery disease
and ventricular tachycardia or fibrillation in the Antiarrhythmics Versus Implantable Defibrillators (AVID) Registry.
Am Heart J. 2002;143:821-6.
2. Velazquez EJ, Lee KL, Jones RH, et al. Coronary-artery bypass surgery in patients with ischemic cardiomyopathy. N
Engl J Med. 2016;374:1511-20.
3. Every NR, Fahrenbruch CE, Hallstrom AP, et al. Influence of coronary bypass surgery on subsequent outcome of
patients resuscitated from out of hospital cardiac arrest. J Am Coll Cardiol. 1992;19:1435-9.
4. Dumas F, Bougouin W, Geri G, et al. Emergency PCI in post-cardiac arrest patients without ST-segment elevation
pattern: insights from the PROCAT II registry. JACC Cardiovasc Interv. 2016;9:1011-8.

4.3.1. Surgery for Arrhythmia Management


Recommendation for Surgery for Arrhythmia Management
References that support the recommendation are summarized in Online Data Supplement 12.
COR LOE Recommendation
1. In patients with monomorphic VT refractory to antiarrhythmic medications
IIb C-LD
and attempts at catheter ablation, surgical ablation may be reasonable (1-7).
References
1. Anter E, Hutchinson MD, Deo R, et al. Surgical ablation of refractory ventricular tachycardia in patients with
nonischemic cardiomyopathy. Circ Arrhythm Electrophysiol. 2011;4:494-500.
2. Bhavani SS, Tchou P, Saliba W, et al. Surgical options for refractory ventricular tachycardia. J Card Surg.
2007;22:533-4.
3. Choi EK, Nagashima K, Lin KY, et al. Surgical cryoablation for ventricular tachyarrhythmia arising from the left
ventricular outflow tract region. Heart Rhythm. 2015;12:1128-36.

Page 34
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

4. Kumar S, Barbhaiya CR, Sobieszczyk P, et al. Role of alternative interventional procedures when endo- and epicardial
catheter ablation attempts for ventricular arrhythmias fail. Circ Arrhythm Electrophysiol. 2015;8:606-15.
5. Mulloy DP, Bhamidipati CM, Stone ML, et al. Cryoablation during left ventricular assist device implantation reduces
postoperative ventricular tachyarrhythmias. J Thorac Cardiovasc Surg. 2013;145:1207-13.
6. Patel M, Rojas F, Shabari FR, et al. Safety and feasibility of open chest epicardial mapping and ablation of ventricular
tachycardia during the period of left ventricular assist device implantation. J Cardiovasc Electrophysiol. 2016;27:95-101.
7. Sartipy U, Albage A, Straat E, et al. Surgery for ventricular tachycardia in patients undergoing left ventricular
reconstruction by the Dor procedure. Ann Thorac Surg. 2006;81:65-71.

4.4. Autonomic Modulation


Recommendations for Autonomic Modulation
References that support the recommendations are summarized in Online Data Supplement 13 and 14.
COR LOE Recommendations
1. In patients with symptomatic, nonlife-threatening VA, treatment with a beta
IIa C-LD
blocker is reasonable (1).
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

2. In patients with VT/VF storm in whom a beta blocker, other antiarrhythmic


IIb C-LD medications, and catheter ablation are ineffective, not tolerated, or not
possible, cardiac sympathetic denervation may be reasonable (2-4).
References
1. Krittayaphong R, Bhuripanyo K, Punlee K, et al. Effect of atenolol on symptomatic ventricular arrhythmia without
structural heart disease: a randomized placebo-controlled study. Am Heart J. 2002;144:e10.
2. Vaseghi M, Barwad P, Malavassi Corrales FJ, et al. Cardiac sympathetic denervation for refractory ventricular
arrhythmias. J Am Coll Cardiol. 2017;69:3070-80.
3. Vaseghi M, Gima J, Kanaan C, et al. Cardiac sympathetic denervation in patients with refractory ventricular
arrhythmias or electrical storm: intermediate and long-term follow-up. Heart Rhythm. 2014;11:360-6.
4. Schwartz PJ, Motolese M, Pollavini G. Prevention of sudden cardiac death after a first myocardial infarction by
pharmacologic or surgical antiadrenergic interventions. J Cardiovasc Electrophysiol. 1992;3:2-16.

Page 35
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

5. Acute Management of Specific VA


Recommendations for Management of Cardiac Arrest
References that support the recommendations are summarized in Online Data Supplement 15 and 16.
COR LOE Recommendations
1. CPR should be performed in patients in cardiac arrest. according to published
I A
basic and advanced cardiovascular life support algorithms (1-3).
2. In patients with hemodynamically unstable VA that persist or recur after a
I A maximal energy shock, intravenous amiodarone should be administered to
attempt to achieve a stable rhythm after further defibrillation (1, 4-6).
3. Patients presenting with VA with hemodynamic instability should undergo
I A
direct current cardioversion (1-3).
4. In patients with polymorphic VT or VF with ST-elevation MI, angiography with
I B-NR
emergency revascularization is recommended (7-10).
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

5. Patients with a wide-QRS tachycardia should be presumed to have VT if the


I C-EO
diagnosis is unclear.
6. In patients with hemodynamically stable VT, administration of intravenous
IIa A
procainamide can be useful to attempt to terminate VT (11-13).
7. In patients with a witnessed cardiac arrest due to VF or polymorphic VT that is
IIa B-R unresponsive to CPR, defibrillation, and vasopressor therapy, intravenous
lidocaine can be beneficial (1, 4, 5, 14, 15).
8. In patients with polymorphic VT due to myocardial ischemia, intravenous beta
IIa B-R
blockers can be useful (16, 17).
9. In patients with a recent MI who have VT/VF that repeatedly recurs despite
IIa B-NR direct current cardioversion and antiarrhythmic medications (VT/VF storm), an
intravenous beta blocker can be useful (17, 18).
10. In patients in cardiac arrest, administration of epinephrine (1 mg every 3 to 5
IIb A
minutes) during CPR may be reasonable (1, 19-24).
11. In patients with hemodynamically stable VT, administration of intravenous
IIb B-R amiodarone or sotalol may be considered to attempt to terminate VT (5, 13,
25, 26).
III: No 12. In patients with cardiac arrest, administration of high-dose epinephrine (>1 mg
A
Benefit boluses) compared with standard doses is not beneficial (19, 21).
III: No 13. In patients with refractory VF not related to torsades de pointes,
A
Benefit administration of intravenous magnesium is not beneficial (27, 28).
14. In patients with suspected AMI, prophylactic administration of lidocaine or
III: Harm B-R
high-dose amiodarone for the prevention of VT is potentially harmful (16, 29).
15. In patients with a wide QRS complex tachycardia of unknown origin, calcium
III: Harm C-LD channel blockers (e.g., verapamil and diltiazem) are potentially harmful (30,
31).
Figure 2

Page 36
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 2. Management of Sustained Monomorphic VT


Sustained
Monomorphic VT

Direct current
Hemodynamic
Stable Unstable cardioversion &
stablility
ACLS

12-lead ECG,
history & physical

Consider disease Cardioversion


specific VTs (Class I)
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Structural IV procainamide VT
Yes
heart disease (Class IIa) termination

No IV amiodarone or
sotalol Yes No
(Class IIb)
Typical ECG
morphology for Therapy guided
by underlying Cardioversion
idiopathic VA
heart disease (Class I)

Verapamil sensitive VT* : verapamil VT


or Yes
termination
Outflow tract VT: beta blocker
for acute termination of VT
Yes
(Class IIa) No

Sedation/anesthesia,
Cardioversion VT reassess antiarrhythmic
Effective No termination therapeutic options,
(Class I)
repeat cardioversion

Yes No

Therapy to prevent Catheter ablation


recurrence preferred (Class I)

Verapamil or
Catheter ablation beta blocker
(Class I) (Class IIa)
Colors correspond to Class of Recommendation in Table 1.
See Sections 7, 8.1.3, 8.2.3, and 10 in the full-text guideline for discussion.
*Known history of verapamil sensitive or classical electrocardiographic presentation.
ACLS indicates advanced cardiovascular life support; ECG, electrocardiogram; VA, ventricular arrhythmia; and VT,
ventricular tachycardia.

Page 37
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

References
1. Link MS, Berkow LC, Kudenchuk PJ, et al. Part 7: adult advanced cardiovascular life support: 2015 American Heart
Association guidelines update for cardiopulmonary resuscitation and emergency cardiovascular care. Circulation.
2015;132(suppl 2):S444-64.
2. Stiell IG, Wells GA, Field B, et al. Advanced cardiac life support in out-of-hospital cardiac arrest. N Engl J Med.
2004;351:647-56.
3. Sasson C, Rogers MA, Dahl J, et al. Predictors of survival from out-of-hospital cardiac arrest: a systematic review and
meta-analysis. Circ Cardiovasc Qual Outcomes. 2010;3:63-81.
4. Dorian P, Cass D, Schwartz B, et al. Amiodarone as compared with lidocaine for shock-resistant ventricular
fibrillation. N Engl J Med. 2002;346:884-90.
5. Kudenchuk PJ, Brown SP, Daya M, et al. Amiodarone, lidocaine, or placebo in out-of-hospital cardiac arrest. N Engl J
Med. 2016;374:1711-22.
6. Kudenchuk PJ, Cobb LA, Copass MK, et al. Amiodarone for resuscitation after out-of-hospital cardiac arrest due to
ventricular fibrillation. N Engl J Med. 1999;341:871-8.
7. Spaulding CM, Joly LM, Rosenberg A, et al. Immediate coronary angiography in survivors of out-of-hospital cardiac
arrest. N Engl J Med. 1997;336:1629-33.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

8. Cronier P, Vignon P, Bouferrache K, et al. Impact of routine percutaneous coronary intervention after out-of-hospital
cardiac arrest due to ventricular fibrillation. Crit Care. 2011;15:R122.
9. Zanuttini D, Armellini I, Nucifora G, et al. Impact of emergency coronary angiography on in-hospital outcome of
unconscious survivors after out-of-hospital cardiac arrest. Am J Cardiol. 2012;110:1723-8.
10.Dumas F, Bougouin W, Geri G, et al. Emergency PCI in post-cardiac arrest patients without ST-segment elevation
pattern: insights from the PROCAT II registry. JACC Cardiovasc Interv. 2016;9:1011-8.
11.Gorgels AP, van den Dool A, Hofs A, et al. Comparison of procainamide and lidocaine in terminating sustained
monomorphic ventricular tachycardia. Am J Cardiol. 1996;78:43-6.
12.Markel DT, Gold LS, Allen J, et al. Procainamide and survival in ventricular fibrillation out-of-hospital cardiac arrest.
Acad Emerg Med. 2010;17:617-23.
13.Ortiz M, Martin A, Arribas F, et al. Randomized comparison of intravenous procainamide vs. intravenous
amiodarone for the acute treatment of tolerated wide QRS tachycardia: the PROCAMIO study. Eur Heart J.
2017;38:1329-35.
14.Herlitz J, Ekstrom L, Wennerblom B, et al. Lidocaine in out-of-hospital ventricular fibrillation. Does it improve
survival? Resuscitation. 1997;33:199-205.
15.Kudenchuk PJ, Newell C, White L, et al. Prophylactic lidocaine for post resuscitation care of patients with out-of-
hospital ventricular fibrillation cardiac arrest. Resuscitation. 2013;84:1512-8.
16.Teo KK, Yusuf S, Furberg CD. Effects of prophylactic antiarrhythmic drug therapy in acute myocardial infarction. An
overview of results from randomized controlled trials. JAMA. 1993;270:1589-95.
17.Nademanee K, Taylor R, Bailey WE, et al. Treating electrical storm : sympathetic blockade versus advanced cardiac
life support-guided therapy. Circulation. 2000;102:742-7.
18.Piccini JP, Hranitzky PM, Kilaru R, et al. Relation of mortality to failure to prescribe beta blockers acutely in patients
with sustained ventricular tachycardia and ventricular fibrillation following acute myocardial infarction (from the
VALsartan In Acute myocardial iNfarcTion trial [VALIANT] Registry). Am J Cardiol. 2008;102:1427-32.
19.Callaham M, Madsen CD, Barton CW, et al. A randomized clinical trial of high-dose epinephrine and norepinephrine
vs standard-dose epinephrine in prehospital cardiac arrest. JAMA. 1992;268:2667-72.
20.Donnino MW, Salciccioli JD, Howell MD, et al. Time to administration of epinephrine and outcome after in-hospital
cardiac arrest with non-shockable rhythms: retrospective analysis of large in-hospital data registry. BMJ.
2014;348:g3028.
21.Gueugniaud PY, Mols P, Goldstein P, et al. A comparison of repeated high doses and repeated standard doses of
epinephrine for cardiac arrest outside the hospital. European Epinephrine Study Group. N Engl J Med.
1998;339:1595-601.
22.Hagihara A, Hasegawa M, Abe T, et al. Prehospital epinephrine use and survival among patients with out-of-hospital
cardiac arrest. JAMA. 2012;307:1161-8.
23.Jacobs IG, Finn JC, Jelinek GA, et al. Effect of adrenaline on survival in out-of-hospital cardiac arrest: a randomised
double-blind placebo-controlled trial. Resuscitation. 2011;82:1138-43.

Page 38
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

24.Koscik C, Pinawin A, McGovern H, et al. Rapid epinephrine administration improves early outcomes in out-of-
hospital cardiac arrest. Resuscitation. 2013;84:915-20.
25.Ho DS, Zecchin RP, Richards DA, et al. Double-blind trial of lignocaine versus sotalol for acute termination of
spontaneous sustained ventricular tachycardia. Lancet. 1994;344:18-23.
26.Somberg JC, Bailin SJ, Haffajee CI, et al. Intravenous lidocaine versus intravenous amiodarone (in a new aqueous
formulation) for incessant ventricular tachycardia. Am J Cardiol 2002;90:853-9.
27.Hassan TB, Jagger C, Barnett DB. A randomised trial to investigate the efficacy of magnesium sulphate for refractory
ventricular fibrillation. Emerg Med J. 2002;19:57-62.
28.Thel MC, Armstrong AL, McNulty SE, et al. Randomised trial of magnesium in in-hospital cardiac arrest. Duke
Internal Medicine Housestaff. Lancet. 1997;350:1272-6.
29.Elizari MV, Martinez JM, Belziti C, et al. Morbidity and mortality following early administration of amiodarone in
acute myocardial infarction. GEMICA study investigators, GEMA Group, Buenos Aires, Argentina. Grupo de Estudios
Multicentricos en Argentina. Eur Heart J. 2000;21:198-205.
30.Belhassen B, Horowitz LN. Use of intravenous verapamil for ventricular tachycardia. Am J Cardiol. 1984;54:1131-3.
31.Buxton AE, Marchlinski FE, Doherty JU, et al. Hazards of intravenous verapamil for sustained ventricular tachycardia.
Am J Cardiol. 1987;59:1107-10.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

6. Ongoing Management of VA and SCD Risk Related to Specific Disease


States
6.1. Ischemic Heart Disease
6.1.1. Secondary Prevention of SCD in Patients With Ischemic Heart Disease
Recommendations for Secondary Prevention of SCD in Patients With Ischemic Heart Disease
References that support the recommendations are summarized in Online Data Supplement 17 and 18.
COR LOE Recommendations
1. In patients with ischemic heart disease, who either survive SCA due to VT/VF or
B-R
experience hemodynamically unstable VT (LOE: B-R) (1-4) or stable VT (LOE: B-
I
NR) (5) not due to reversible causes, an ICD is recommended if meaningful
B-NR
survival greater than 1 year is expected.
Value 2. A transvenous ICD provides intermediate value in the secondary prevention of
Statement: SCD particularly when the patients risk of death due to a VA is deemed high and
Intermediate the risk of nonarrhythmic death (either cardiac or noncardiac) is deemed low
Value
based on the patients burden of comorbidities and functional status (6).
(LOE: B-R)
3. In patients with ischemic heart disease and unexplained syncope who have
I B-NR inducible sustained monomorphic VT on electrophysiological study, an ICD is
recommended if meaningful survival of greater than 1 year is expected (7).
Figure 3

Page 39
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 3. Secondary Prevention Patients With Ischemic Heart Disease


Secondary prevention in
pts with IHD

SCA survivor*
or sustained Cardiac syncope
spontaneous
monomorphic VT*
LVEF35%

Ischemia Yes No
warranting
revascularization ICD EP study
(Class I) (Class IIa)

Yes No
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Revascularize Inducible
& reassess VA
SCD risk ICD candidate
(Class I)
Yes No Yes No

ICD GDMT ICD Extended


(Class I) (Class I) (Class I) monitoring

Colors correspond to Class of Recommendation in Table 1.


See Sections 4.3.1 and 7.1.1 in the full-text guideline for discussion.
*Exclude reversible causes.
History consistent with an arrhythmic etiology for syncope.
ICD candidacy as determined by functional status, life expectancy, or patient preference.
EP indicates electrophysiological; GDMT, guideline-directed management and therapy; ICD, implantable cardioverter-
defibrillator; IHD, ischemic heart disease; LVEF, left ventricular ejection fraction; pts, patients; SCA, sudden cardiac arrest;
SCD, sudden cardiac death; and VT, ventricular tachycardia.

References
1. The AVID Investigators. A comparison of antiarrhythmic-drug therapy with implantable defibrillators in patients
resuscitated from near-fatal ventricular arrhythmias. N Engl J Med. 1997;337:1576-83.
2. Connolly SJ, Gent M, Roberts RS, et al. Canadian implantable defibrillator study (CIDS) : a randomized trial of the
implantable cardioverter defibrillator against amiodarone. Circulation. 2000;101:1297-302.
3. Connolly SJ, Hallstrom AP, Cappato R, et al. Meta-analysis of the implantable cardioverter defibrillator secondary
prevention trials. AVID, CASH and CIDS studies. Antiarrhythmics vs Implantable Defibrillator study. Cardiac Arrest
Study Hamburg . Canadian Implantable Defibrillator Study. Eur Heart J. 2000;21:2071-8.
4. Kuck KH, Cappato R, Siebels J, et al. Randomized comparison of antiarrhythmic drug therapy with implantable
defibrillators in patients resuscitated from cardiac arrest: the Cardiac Arrest Study Hamburg (CASH). Circulation.
2000;102:748-54.
5. Raitt MH, Renfroe EG, Epstein AE, et al. "Stable" ventricular tachycardia is not a benign rhythm : insights from the
antiarrhythmics versus implantable defibrillators (AVID) registry. Circulation. 2001;103:244-52.
6. Owens DK, Sanders GD, Heidenreich PA, et al. Effect of risk stratification on cost-effectiveness of the implantable
cardioverter defibrillator. Am Heart J. 2002;144:440-8.
7. Bass EB, Elson JJ, Fogoros RN, et al. Long-term prognosis of patients undergoing electrophysiologic studies for
syncope of unknown origin. Am J Cardiol. 1988;62:1186-91.

Page 40
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.1.1.1. Coronary Artery Spasm


Recommendations for Patients With Coronary Artery Spasm
References that support the recommendations are summarized in Online Data Supplement 20.
COR LOE Recommendations
1. In patients with VA due to coronary artery spasm, treatment with maximally
I B-NR tolerated doses of a calcium channel blocker and smoking cessation are
indicated to reduce recurrent ischemia and VA (1, 2).
2. In patients resuscitated from SCA due to coronary artery spasm in whom
IIa B-NR medical therapy is ineffective or not tolerated, an ICD is reasonable if
meaningful survival of greater than 1 year is expected (3-6).
3. In patients resuscitated from SCA due to coronary artery spasm, an ICD in
IIb B-NR addition to medical therapy may be reasonable if meaningful survival of
greater than 1 year is expected (3-6).
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

References
1. Chevalier P, Dacosta A, Defaye P, et al. Arrhythmic cardiac arrest due to isolated coronary artery spasm: long-term
outcome of seven resuscitated patients. J Am Coll Cardiol. 1998;31:57-61.
2. Myerburg RJ, Kessler KM, Mallon SM, et al. Life-threatening ventricular arrhythmias in patients with silent
myocardial ischemia due to coronary-artery spasm. N Engl J Med. 1992;326:1451-5.
3. Ahn JM, Lee KH, Yoo SY, et al. Prognosis of variant angina manifesting as aborted sudden cardiac death. J Am Coll
Cardiol. 2016;68:137-45.
4. Matsue Y, Suzuki M, Nishizaki M, et al. Clinical implications of an implantable cardioverter-defibrillator in patients
with vasospastic angina and lethal ventricular arrhythmia. J Am Coll Cardiol. 2012;60:908-13.
5. Takagi Y, Yasuda S, Tsunoda R, et al. Clinical characteristics and long-term prognosis of vasospastic angina patients
who survived out-of-hospital cardiac arrest: multicenter registry study of the Japanese Coronary Spasm Association.
Circ Arrhythm Electrophysiol. 2011;4:295-302.
6. Meisel SR, Mazur A, Chetboun I, et al. Usefulness of implantable cardioverter-defibrillators in refractory variant
angina pectoris complicated by ventricular fibrillation in patients with angiographically normal coronary arteries. Am
J Cardiol. 2002;89:1114-6.

Page 41
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.1.2. Primary Prevention of SCD in Patients With Ischemic Heart Disease


Recommendations for Primary Prevention of SCD in Patients With Ischemic Heart Disease
References that support the recommendations are summarized in Online Data Supplement 21.
COR LOE Recommendations
1. In patients with LVEF of 35% or less that is due to ischemic heart disease who
are at least 40 days post-MI and at least 90 days postrevascularization, and
I A
with NYHA class II or III HF despite GDMT, an ICD is recommended if meaningful
survival of greater than 1 year is expected (1, 2).
2. In patients with LVEF of 30% or less that is due to ischemic heart disease who
are at least 40 days post-MI and at least 90 days postrevascularization, and
I A
with NYHA class I HF despite GDMT, an ICD is recommended if meaningful
survival of greater than 1 year is expected (2, 3).
3. A transvenous ICD provides high value in the primary prevention of SCD
Value Statement:
particularly when the patients risk of death due to a VA is deemed high and
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

High Value
the risk of nonarrhythmic death (either cardiac or noncardiac) is deemed low
(LOE: B-R)
based on the patients burden of comorbidities and functional status (4).
4. In patients with NSVT due to prior MI, LVEF of 40% or less and inducible
I B-R sustained VT or VF at electrophysiological study, an ICD is recommended if
meaningful survival of greater than 1 year is expected (5).
5. In nonhospitalized patients with NYHA class IV symptoms who are candidates
IIa B-NR for cardiac transplantation or an LVAD, an ICD is reasonable if meaningful
survival of greater than 1 year is expected (6-9).
6. An ICD is not indicated for NYHA class IV patients with medication-refractory
III: No
C-EO HF who are not also candidates for cardiac transplantation, an LVAD, or a CRT
Benefit
defibrillator that incorporates both pacing and defibrillation capabilities.
Figure 4

Page 42
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 4. Primary Prevention of SCD in Patients With Ischemic Heart Disease


Primary prevention in pts with IHD,
EP study
LVEF 40%
(especially in the Inducible ICD
Yes
presence of sustained VT (Class I)
NSVT)
MI <40 d
and/or GDMT
Yes* No
revascularization (Class I)
<90 d
Reassess LVEF
WCD >40 d after MI
(Class IIb) and/or >90 d after
No revascularization

NYHA NYHA
NYHA class I LVEF 40%,
class II or III class IV candidate
LVEF 30% NSVT, inducible
LVEF 35% for advanced HF
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

sustained VT on
therapy
EP study

Yes Yes No Yes No

ICD ICD ICD ICD should not


GDMT be implanted
(Class I)* (Class I) (Class IIa)
(Class III:
No Benefit)
Colors correspond to Class of Recommendation in Table 1.
See Section 7.1.2 in the full-text guideline for discussion.
*Scenarios exist for early ICD placement in select circumstances such as patients with a pacing indication or syncope.
Advanced HF therapy includes CRT, cardiac transplant, and LVAD.
thought due to VT. These are detailed elsewhere in an HRS/ACC/AHA expert consensus statement (24).
CRT indicates cardiac resynchronization therapy; EP, electrophysiological; GDMT, guideline-directed management and
therapy; HF, heart failure; ICD, implantable cardioverter-defibrillator; IHD, ischemic heart disease; LVEF, left ventricular
ejection fraction; MI, myocardial infarction; NSVT, nonsustained ventricular tachycardia; NYHA, New York Heart
Association; pts, patients; SCD, sudden cardiac death; VT, ventricular tachycardia; and WCD, wearable cardioverter-
defibrillator.

References
1. Bardy GH, Lee KL, Mark DB, et al. Amiodarone or an implantable cardioverter-defibrillator for congestive heart
failure. N Engl J Med. 2005;352:225-37.
2. Moss AJ, Zareba W, Hall WJ, et al. Prophylactic implantation of a defibrillator in patients with myocardial infarction
and reduced ejection fraction. N Engl J Med. 2002;346:877-83.
3. Moss AJ, Hall WJ, Cannom DS, et al. Improved survival with an implanted defibrillator in patients with coronary
disease at high risk for ventricular arrhythmia. Multicenter Automatic Defibrillator Implantation Trial Investigators.
N Engl J Med. 1996;335:1933-40.
4. Owens DK, Sanders GD, Heidenreich PA, et al. Effect of risk stratification on cost-effectiveness of the implantable
cardioverter defibrillator. Am Heart J. 2002;144:440-8.
5. Buxton AE, Lee KL, Fisher JD, et al. A randomized study of the prevention of sudden death in patients with coronary
artery disease. Multicenter Unsustained Tachycardia Trial Investigators. N Engl J Med. 1999;341:1882-90.
6. Cantero-Prez EM, Sobrino-Mrquez JM, Grande-Trillo A, et al. Implantable cardioverter defibrillator for primary
prevention in patients with severe ventricular dysfunction awaiting heart transplantation. Transplant Proc.
2013;45:3659-61.
7. Frohlich GM, Holzmeister J, Hubler M, et al. Prophylactic implantable cardioverter defibrillator treatment in
patients with end-stage heart failure awaiting heart transplantation. Heart. 2013;99:1158-65.

Page 43
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

8. Gandjbakhch E, Rovani M, Varnous S, et al. Implantable cardioverter-defibrillators in end-stage heart failure


patients listed for heart transplantation: results from a large retrospective registry. Arch Cardiovasc Diseases.
2016;109:476-85.
9. Vakil K, Duval S, Cogswell R, et al. Impact of implantable cardioverter-defibrillators on waitlist mortality among
patients awaiting heart transplantation: an UNOS/OPTN analysis. JACC Clin Electrophysiol. 2017;3:33-40.

6.1.3. Treatment and Prevention of Recurrent VA in Patients With Ischemic Heart


Disease
Recommendations for Treatment of Recurrent VA in Patients With Ischemic Heart Disease
References that support the recommendations are summarized in Online Data Supplement 22 and 23.
COR LOE Recommendations
1. In patients with ischemic heart disease and recurrent VA, with significant
symptoms or ICD shocks despite optimal device programming and ongoing
I B-R
treatment with a beta blocker, amiodarone or sotalol is useful to suppress
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

recurrent VA (1-3).
2. In patients with prior MI and recurrent episodes of symptomatic sustained VT,
B-R or who present with VT or VF storm and have failed or are intolerant of
I amiodarone (LOE: B-R) (4) or other antiarrhythmic medications (LOE: B-NR) (5-
B-NR 9), catheter ablation is recommended (10-12).
3. In patients with ischemic heart disease and ICD shocks for sustained
monomorphic VT or symptomatic sustained monomorphic VT that is
IIb C-LD
recurrent, or hemodynamically tolerated, catheter ablation as first-line
therapy may be considered to reduce recurrent VA (10, 11).
III: 4. In patients with prior MI, class IC antiarrhythmic medications (e.g., flecainide
B-R
Harm and propafenone) should not be used (13).
III: 5. In patients with incessant VT or VF, an ICD should not be implanted until
C-LD
Harm sufficient control of the VA is achieved to prevent repeated ICD shocks (14).
6. In patients with ischemic heart disease and sustained monomorphic VT,
III: No
C-LD coronary revascularization alone is an ineffective therapy to prevent recurrent
Benefit
VT (15, 16).
Figure 5

Page 44
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 5. Treatment of Recurrent VA in Patients With Ischemic Heart Disease or NICM


ICD with VT/VF
recurrent arrhythmia*

Polymorphic Sustained
VT/VF monomorphic VT

Consider
Catheter ablation Amiodarone or
reversible
as first-line therapy sotalol
causes
(Class IIb) (Class I)

Drug, electrolyte induced Ischemia No reversible causes Arrhythmia


not controlled
Treat for QT prolongation, Revascularize Amiodarone Beta blockers
discontinue offending (Class I) (Class I) or lidocaine
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

medication, (Class IIa)


correct electrolytes
(Class I) IHD with NICM
frequent VT or
Arrhythmia VT storm
not controlled
Yes No

Catheter Catheter Catheter


ablation ablation ablation
Identifiable
(Class I) (Class IIa) (Class IIa)
PVC triggers

Yes No

Catheter Autonomic
ablation modulation
(Class I) (Class IIb)
Colors correspond to Class of Recommendation in Table 1.
See Sections 5.6, 6, 7.1.3, and 7.2 in the full-text guideline for discussion.
*Management should start with ensuring that the ICD is programmed appropriately and that potential precipitating
causes, including heart failure exacerbation, are addressed. For information regarding optimal ICD programming, refer
to the 2015 HRS/EHRA/APHRS/SOLAECE expert consensus statement (26).
EHRA indicated European Heart Rhythm Association; HRS, Heart Rhythm Society; IHD, ischemic heart disease; ICD,
implantable cardioverter-defibrillator; PVC, premature ventricular complex; NICM, nonischemic cardiomyopathy;
SOLAECE, Sociedad Latinoamericana de Estimulacin Cardaca y Electrosiologa; VF, ventricular fibrillation; and VT,
ventricular tachycardia.

References
1. Connolly SJ, Dorian P, Roberts RS, et al. Comparison of beta-blockers, amiodarone plus beta-blockers, or sotalol for
prevention of shocks from implantable cardioverter defibrillators: the OPTIC Study: a randomized trial. JAMA.
2006;295:165-71.
2. Pacifico A, Hohnloser SH, Williams JH, et al. Prevention of implantable-defibrillator shocks by treatment with sotalol.
d,l-Sotalol Implantable Cardioverter-Defibrillator Study Group. N Engl J Med. 1999;340:1855-62.
3. Kettering K, Mewis C, Dornberger V, et al. Efficacy of metoprolol and sotalol in the prevention of recurrences of
sustained ventricular tachyarrhythmias in patients with an implantable cardioverter defibrillator. Pacing Clin
Electrophysiol. 2002;25:1571-6.
4. Sapp JL, Wells GA, Parkash R, et al. Ventricular tachycardia ablation versus escalation of antiarrhythmic drugs. N Engl
J Med. 2016;375:111-21.

Page 45
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

5. Mallidi J, Nadkarni GN, Berger RD, et al. Meta-analysis of catheter ablation as an adjunct to medical therapy for
treatment of ventricular tachycardia in patients with structural heart disease. Heart Rhythm. 2011;8:503-10.
6. Marchlinski FE, Haffajee CI, Beshai JF, et al. Long-term success of irrigated radiofrequency catheter ablation of
sustained ventricular tachycardia: post-approval THERMOCOOL VT trial. J Am Coll Cardiol. 2016;67:674-83.
7. Stevenson WG, Wilber DJ, Natale A, et al. Irrigated radiofrequency catheter ablation guided by electroanatomic
mapping for recurrent ventricular tachycardia after myocardial infarction: the multicenter thermocool ventricular
tachycardia ablation trial. Circulation. 2008;118:2773-82.
8. Tanner H, Hindricks G, Volkmer M, et al. Catheter ablation of recurrent scar-related ventricular tachycardia using
electroanatomical mapping and irrigated ablation technology: results of the prospective multicenter Euro-VT-study.
J Cardiovasc Electrophysiol. 2010;21:47-53.
9. Tung R, Vaseghi M, Frankel DS, et al. Freedom from recurrent ventricular tachycardia after catheter ablation is
associated with improved survival in patients with structural heart disease: an International VT Ablation Center
Collaborative Group study. Heart Rhythm. 2015;12:1997-2007.
10.Al-Khatib SM, Daubert JP, Anstrom KJ, et al. Catheter ablation for ventricular tachycardia in patients with an
implantable cardioverter defibrillator (CALYPSO) pilot trial. J Cardiovasc Electrophysiol. 2015;26:151-7.
11.Kuck KH, Schaumann A, Eckardt L, et al. Catheter ablation of stable ventricular tachycardia before defibrillator
implantation in patients with coronary heart disease (VTACH): a multicentre randomised controlled trial. Lancet.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

2010;375:31-40.
12.Sesselberg HW, Moss AJ, McNitt S, et al. Ventricular arrhythmia storms in postinfarction patients with implantable
defibrillators for primary prevention indications: a MADIT-II substudy. Heart Rhythm. 2007;4:1395-402.
13.Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in patients receiving encainide, flecainide, or
placebo. The Cardiac Arrhythmia Suppression Trial. N Engl J Med. 1991;324:781-8.
14.Sears SF Jr, Todaro JF, Lewis TS, et al. Examining the psychosocial impact of implantable cardioverter defibrillators: a
literature review. Clinical Cardiol. 1999;22:481-9.
15.Steinberg JS, Gaur A, Sciacca R, et al. New-onset sustained ventricular tachycardia after cardiac surgery. Circulation.
1999;99:903-8.
16.Brugada J, Aguinaga L, Mont L, et al. Coronary artery revascularization in patients with sustained ventricular
arrhythmias in the chronic phase of a myocardial infarction: effects on the electrophysiologic substrate and
outcome. J Am Coll Cardiol. 2001;37:529-33.

6.2. Nonischemic Cardiomyopathy


Recommendations for Patients With NICM
References that support the recommendations are summarized in Online Data Supplement 24.
COR LOE Recommendations
1. In patients with suspected NICM from myocardial infiltrative processess,
I B-NR
cardiac MRI with late gadolinium enhancement is useful for diagnosis (1-3).
2. In patients with suspected NICM, cardiac MRI with late gadolinium
IIa B-NR
enhancement can be useful for assessing risk of SCA/SCD (1-3).
3. In patients with NICMwho develop conduction disease or LV dysfunction at
less than 40 years of age, or who have a family history of NICMor SCD in a first-
IIa C-EO degree relative (<50 years of age), genetic counseling and genetic testing are
reasonable to detect a heritable disease that may clarify prognosis and
facilitate cascade screening of relatives (4, 5).

References
1. Greulich S, Deluigi CC, Gloekler S, et al. CMR imaging predicts death and other adverse events in suspected cardiac
sarcoidosis. JACC Cardiovasc Imaging. 2013;6:501-11.
2. Kuruvilla S, Adenaw N, Katwal AB, et al. Late gadolinium enhancement on cardiac magnetic resonance predicts
adverse cardiovascular outcomes in nonischemic cardiomyopathy: a systematic review and meta-analysis. Circ
Cardiovasc Imaging. 2014;7:250-8.

Page 46
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

3. Piers SR, Tao Q, van Huls van Taxis CFB, et al. Contrast-enhanced MRI-derived scar patterns and associated
ventricular tachycardias in nonischemic cardiomyopathy: implications for the ablation strategy. Circ Arrhythm
Electrophysiol. 2013;6:875-83.
4. Ackerman MJ, Priori SG, Willems S, et al. HRS/EHRA expert consensus statement on the state of genetic testing
for the channelopathies and cardiomyopathies: this document was developed as a partnership between the
Heart Rhythm Society (HRS) and the European Heart Rhythm Association (EHRA). Europace. 2011;13:1077-109.
5. Hershberger RE, Morales A, Siegfried JD. Clinical and genetic issues in dilated cardiomyopathy: a review for
genetics professionals. Genet Med. 2010;12:655-67.

6.2.1. Secondary Prevention of SCD in Patients With NICM


Recommendations for Secondary Prevention of SCD in Patients With NICM
References that support the recommendations are summarized in Online Data Supplement 25 and 26.
COR LOE Recommendations
1. In patients with NICM who either survive SCA due to VT/VF or experience
B-R
hemodynamically unstable VT (LOE: B-R) (1-4) or stable VT (LOE: B-NR) (5) not
I
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

due to reversible causes, an ICD is recommended if meaningful survival greater


B-NR
than 1 year is expected.
2. In patients with NICM who experience syncope presumed to be due to VA and
who do not meet indications for a primary prevention ICD, an ICD or an
IIa B-NR
electrophysiological study for risk stratification for SCD can be beneficial if
meaningful survival greater than 1 year is expected (6-11).
3. In patients with NICM who survive a cardiac arrest, have sustained VT, or have
symptomatic VA who are ineligible for an ICD (due to a limited life-expectancy
IIb B-R
and/or functional status or lack of access to an ICD), amiodarone may be
considered for prevention of SCD (12, 13).
Figure 6
References
1. The Antiarrhythmics versus Implantable Defibrillators (AVID) Investigators. A comparison of antiarrhythmic-
drug therapy with implantable defibrillators in patients resuscitated from near-fatal ventricular arrhythmias. N
Engl J Med. 1997;337:1576-83.
2. Connolly SJ, Gent M, Roberts RS, et al. Canadian implantable defibrillator study (CIDS): a randomized trial of the
implantable cardioverter defibrillator against amiodarone. Circulation. 2000;101:1297-302.
3. Desai AS, Fang JC, Maisel WH, et al. Implantable defibrillators for the prevention of mortality in patients with
nonischemic cardiomyopathy: a meta-analysis of randomized controlled trials. JAMA. 2004;292:2874-9.
4. Kuck KH, Cappato R, Siebels J, et al. Randomized comparison of antiarrhythmic drug therapy with implantable
defibrillators in patients resuscitated from cardiac arrest : the Cardiac Arrest Study Hamburg (CASH).
Circulation. 2000;102:748-54.
5. Raitt MH, Renfroe EG, Epstein AE, et al. "Stable" ventricular tachycardia is not a benign rhythm : insights from
the Antiarrhythmics versus Implantable Defibrillators (AVID) registry. Circulation. 2001;103:244-52.
6. Brilakis ES, Shen WK, Hammill SC, et al. Role of programmed ventricular stimulation and implantable
cardioverter defibrillators in patients with idiopathic dilated cardiomyopathy and syncope. Pacing Clin
Electrophysiol. 2001;24:1623-30.
7. Fonarow GC, Feliciano Z, Boyle NG, et al. Improved survival in patients with nonischemic advanced heart failure
and syncope treated with an implantable cardioverter-defibrillator. Am J Cardiol. 2000;85:981-5.
8. Knight BP, Goyal R, Pelosi F, et al. Outcome of patients with nonischemic dilated cardiomyopathy and
unexplained syncope treated with an implantable defibrillator. J Am Coll Cardiol. 1999;33:1964-70.
9. Middlekauff HR, Stevenson WG, Stevenson LW, et al. Syncope in advanced heart failure: high risk of sudden
death regardless of origin of syncope. J Am Coll Cardiol. 1993;21:110-6.

Page 47
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

10.Olshansky B, Poole JE, Johnson G, et al. Syncope predicts the outcome of cardiomyopathy patients: analysis of
the SCD-HeFT study. J Am Coll Cardiol. 2008;51:1277-82.
11.Ruwald MH, Okumura K, Kimura T, et al. Syncope in high-risk cardiomyopathy patients with implantable
defibrillators: frequency, risk factors, mechanisms, and association with mortality: results from the multicenter
automatic defibrillator implantation trial-reduce inappropriate therapy (MADIT-RIT) study. Circulation.
2014;129:545-52.
12.Piccini JP, Berger JS, O'Connor CM. Amiodarone for the prevention of sudden cardiac death: a meta-analysis of
randomized controlled trials. Eur Heart J. 2009;30:1245-53.
13.Claro JC, Candia R, Rada G, et al. Amiodarone versus other pharmacological interventions for prevention of
sudden cardiac death. Cochrane Database Syst Rev. 2015;12:CD008093.

6.2.2. Primary Prevention of SCD in Patients With NICM


Recommendations for Primary Prevention of SCD in Patients With NICM
References that support the recommendations are summarized in Online Data Supplement 27 and 28.
COR LOE Recommendations
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

1. In patients with NICM, HF with NYHA class IIIII symptoms and an LVEF of 35%
I A or less, despite GDMT, an ICD is recommended if meaningful survival of greater
than 1 year is expected (1-6).
2. In patients with NICM due to a Lamin A/C mutation who have 2 or more risk
IIa B-NR factors (NSVT, LVEF <45%, nonmissense mutation, and male sex), an ICD can
be beneficial if meaningful survival of greater than 1 year is expected (7-10).
3. In patients with NICM, HF with NYHA class I symptoms and an LVEF of 35% or
IIb B-R less, despite GDMT, an ICD may be considered if meaningful survival of greater
than 1 year is expected (5).
4. In patients with medication-refractory NYHA class IV HF who are not also
III: No candidates for cardiac transplantation, an LVAD, or a CRT defibrillator that
C-EO
Benefit incorporates both pacing and defibrillation capabilities, an ICD should not be
implanted.
Figure 6

References
1. Bardy GH, Lee KL, Mark DB, et al. Amiodarone or an implantable cardioverter-defibrillator for congestive heart
failure. N Engl J Med. 2005;352:225-37.
2. Bnsch D, Antz M, Boczor S, et al. Primary prevention of sudden cardiac death in idiopathic dilated
cardiomyopathy: the Cardiomyopathy Trial (CAT). Circulation. 2002;105:1453-8.
3. Bristow MR, Saxon LA, Boehmer J, et al. Cardiac-resynchronization therapy with or without an implantable
defibrillator in advanced chronic heart failure. N Engl J Med. 2004;350:2140-50.
4. Desai AS, Fang JC, Maisel WH, et al. Implantable defibrillators for the prevention of mortality in patients with
nonischemic cardiomyopathy: a meta-analysis of randomized controlled trials. JAMA. 2004;292:2874-9.
5. Kadish A, Dyer A, Daubert JP, et al. Prophylactic defibrillator implantation in patients with nonischemic dilated
cardiomyopathy. N Engl J Med. 2004;350:2151-8.
6. Strickberger SA, Hummel JD, Bartlett TG, et al. Amiodarone versus implantable cardioverter-defibrillator:
randomized trial in patients with nonischemic dilated cardiomyopathy and asymptomatic nonsustained
ventricular tachycardia--AMIOVIRT. J Am Coll Cardiol. 2003;41:1707-12.
7. Anselme F, Moubarak G, Savoure A, et al. Implantable cardioverter-defibrillators in lamin A/C mutation carriers
with cardiac conduction disorders. Heart Rhythm. 2013;10:1492-8.
8. Pasotti M, Klersy C, Pilotto A, et al. Long-term outcome and risk stratification in dilated cardiolaminopathies. J
Am Coll Cardiol. 2008;52:1250-60.

Page 48
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

9. Van Berlo JH, de Voogt WG, van der Kooi AJ, et al. Meta-analysis of clinical characteristics of 299 carriers of
LMNA gene mutations: do lamin A/C mutations portend a high risk of sudden death? J Mol Med. 2005;83:79-
83.
10.van Rijsingen IA, Arbustini E, Elliott PM, et al. Risk factors for malignant ventricular arrhythmias in lamin A/C
mutation carriers a European cohort study. J Am Coll Cardiol. 2012;59:493-500.

6.2.3. Treatment of Recurrent VA in Patients With NICM


Recommendations for Treatment of Recurrent VA in Patients With NICM
References that support the recommendations are summarized in Online Data Supplement 29.
COR LOE Recommendations
1. In patients with NICM and an ICD who experience spontaneous VA or recurrent
IIa B-R appropriate shocks despite optimal device programming and treatment with
a beta blocker, amiodarone or sotalol can be beneficial (1).
2. In patients with NICM and recurrent sustained monomorphic VT who fail or
IIa B-NR are intolerant of antiarrhythmic medications, catheter ablation can be useful
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

for reducing recurrent VT and ICD shocks (2, 3).

Page 49
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 6. Secondary and Primary Prevention of SCD in Patients With NICM

Patients with NICM

SCA survivor/ NICM due to


Symptoms Class II-III
sustained VT LMNA mutation
No concerning No HF and No
(spontaneous/ and 2 risk
for VA LVEF 35%
inducible) factors

Yes Yes
Yes
Yes

Arrythmogenic ICD ICD


ICD candidate* candidate*
syncope
candidate*
suspected
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Yes No, due to newly Yes


Yes No Yes Etiology uncertain diagnosed HF
(<3 mo GDMT)
ICD Amiodarone ICD EP Study ICD or not on optimal ICD
(Class I) (Class IIb) (Class IIa) (Class IIa) (Class I) GDMT (Class IIa)
If positive
If LVEF 35%
and WCD
Class II-III (Class IIb)
HF
Reassess
LVEF 3mo
Colors correspond to Class of Recommendation in Table 1.
See Section 7.2 in the full-text guideline for discussion.
*ICD candidacy as determined by functional status, life expectancy or patient preference.
2 indicates secondary; EP, electrophysiological; GDMT, guideline-directed management and therapy; HF, heart
failure; ICD, implantable cardioverter-defibrillator; LVEF, left ventricular ejection fraction; NICM, nonischemic
cardiomyopathy; SCA, sudden cardiac arrest; SCD, sudden cardiac death; VA, ventricular arrhythmia; and WCD,
wearable cardiac-defibrillator.

References
1. Connolly SJ, Dorian P, Roberts RS, et al. Comparison of beta-blockers, amiodarone plus beta-blockers, or
sotalol for prevention of shocks from implantable cardioverter defibrillators: the OPTIC Study: a randomized
trial. JAMA. 2006;295:165-71.
2. Dinov B, Fiedler L, Schonbauer R, et al. Outcomes in catheter ablation of ventricular tachycardia in dilated
nonischemic cardiomyopathy compared with ischemic cardiomyopathy: results from the Prospective Heart
Centre of Leipzig VT (HELP-VT) Study. Circulation. 2014;129:728-36.
3. Tung R, Vaseghi M, Frankel DS, et al. Freedom from recurrent ventricular tachycardia after catheter ablation is
associated with improved survival in patients with structural heart disease: an International VT Ablation
Center Collaborative Group study. Heart Rhythm. 2015;12:1997-2007.

Page 50
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.3. Arrhythmogenic Right Ventricular Cardiomyopathy


Recommendations for Arrhythmogenic Right Ventricular Cardiomyopathy
References that support the recommendations are summarized in Online Data Supplement 30.
COR LOE Recommendations
1. In selected first-degree relatives of patients with arrhythmogenic right
ventricular cardiomyopathy, clinical screening for the disease is recommended
I B-NR
along with genetic counseling and genetic testing, if the proband has a disease
causing mutation (1-4).
2. In patients with suspected arrhythmogenic right ventricular cardiomyopathy
I B-NR and VA or electrocardiographic abnormalities, cardiac MRI is useful for
establishing a diagnosis and for risk stratification (5-8).
3. In patients with arrhythmogenic right ventricular cardiomyopathy and an
additional marker of increased risk of SCD (resuscitated SCA, sustained VT,
I B-NR
significant ventricular dysfunction with RVEF or LVEF 35%), an ICD is
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

recommended if meaningful survival greater than 1 year is expected (9-13).


4. In patients with arrhythmogenic right ventricular cardiomyopathy and VA, a
I B-NR
beta blocker is recommended (11, 14, 15).
5. In patients with a clinical diagnosis of arrhythmogenic right ventricular
I B-NR
cardiomyopathy , avoiding intensive exercise is recommended (11, 12, 16-21).
6. In patients with clinically diagnosed or suspected arrhythmogenic right
ventricular cardiomyopathy , genetic counseling and genetic testing can be
IIa B-NR
useful for diagnosis and for gene-specific targeted family screening (1, 4, 22-
26).
7. In patients with arrhythmogenic right ventricular cardiomyopathy and
IIa B-NR syncope presumed due to VA, an ICD can be useful if meaningful survival
greater than 1 year is expected (10, 11, 13).
8. In patients with clinical evidence of arrhythmogenic right ventricular
IIa B-NR
cardiomyopathy but not VA, a beta blocker can be useful (14, 15).
9. In patients with arrhythmogenic right ventricular cardiomyopathy and
recurrent symptomatic sustained VT in whom a beta blocker is ineffective or
IIa B-NR
not tolerated, catheter ablation with availability of a combined
endocardial/epicardial approach can be beneficial (27-33).
10. In patients with suspected arrhythmogenic right ventricular cardiomyopathy ,
IIa B-NR a signal averaged ECG can be useful for diagnosis and risk stratification (14, 34,
35).
11. In asymptomatic patients with clinical evidence of arrhythmogenic right
IIb B-NR ventricular cardiomyopathy , an electrophysiological study may be considered
for risk stratification (9, 36).

References
1. Groeneweg JA, Bhonsale A, James CA, et al. Clinical presentation, long-term follow-up, and outcomes of 1001
arrhythmogenic right ventricular dysplasia/cardiomyopathy patients and family members. Circ Cardiovasc
Genet. 2015;8:437-46.
2. Marcus FI, Edson S, Towbin JA. Genetics of arrhythmogenic right ventricular cardiomyopathy: a practical guide
for physicians. J Am Coll Cardiol. 2013;61:1945-8.
3. Quarta G, Muir A, Pantazis A, et al. Familial evaluation in arrhythmogenic right ventricular cardiomyopathy:
impact of genetics and revised task force criteria. Circulation. 2011;123:2701-9.

Page 51
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

4. te Riele AS, James CA, Groeneweg JA, et al. Approach to family screening in arrhythmogenic right ventricular
dysplasia/cardiomyopathy. Eur Heart J. 2016;37:755-63.
5. Liu T, Pursnani A, Sharma UC, et al. Effect of the 2010 task force criteria on reclassification of cardiovascular
magnetic resonance criteria for arrhythmogenic right ventricular cardiomyopathy. J Cardiovasc Magn Reson.
2014;16:47.
6. Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic right ventricular
cardiomyopathy/dysplasia: proposed modification of the task force criteria. Circulation. 2010;121:1533-41.
7. te Riele AS, Bhonsale A, James CA, et al. Incremental value of cardiac magnetic resonance imaging in
arrhythmic risk stratification of arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated
desmosomal mutation carriers. J Am Coll Cardiol. 2013;62:1761-9.
8. Vermes E, Strohm O, Otmani A, et al. Impact of the revision of arrhythmogenic right ventricular
cardiomyopathy/dysplasia task force criteria on its prevalence by CMR criteria. JACC Cardiovasc Imag.
2011;4:282-7.
9. Bhonsale A, James CA, Tichnell C, et al. Incidence and predictors of implantable cardioverter-defibrillator
therapy in patients with arrhythmogenic right ventricular dysplasia/cardiomyopathy undergoing implantable
cardioverter-defibrillator implantation for primary prevention. J Am Coll Cardiol. 2011;58:1485-96.
10. Corrado D, Calkins H, Link MS, et al. Prophylactic implantable defibrillator in patients with arrhythmogenic
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

right ventricular cardiomyopathy/dysplasia and no prior ventricular fibrillation or sustained ventricular


tachycardia. Circulation. 2010;122:1144-52.
11. Corrado D, Wichter T, Link MS, et al. Treatment of arrhythmogenic right ventricular cardiomyopathy/dysplasia:
an International Task Force Consensus Statement. Circulation. 2015;132:441-53.
12. Link MS, Laidlaw D, Polonsky B, et al. Ventricular arrhythmias in the North American multidisciplinary study of
ARVC: predictors, characteristics, and treatment. J Am Coll Cardiol. 2014;64:119-25.
13. Piccini JP, Dalal D, Roguin A, et al. Predictors of appropriate implantable defibrillator therapies in patients with
arrhythmogenic right ventricular dysplasia. Heart Rhythm. 2005;2:1188-94.
14. Marcus FI, Zareba W, Calkins H, et al. Arrhythmogenic right ventricular cardiomyopathy/dysplasia clinical
presentation and diagnostic evaluation: results from the North American Multidisciplinary Study. Heart
Rhythm. 2009;6:984-92.
15. Marcus GM, Glidden DV, Polonsky B, et al. Efficacy of antiarrhythmic drugs in arrhythmogenic right ventricular
cardiomyopathy: a report from the North American ARVC Registry. J Am Coll Cardiol. 2009;54:609-15.
16. Corrado D, Basso C, Thiene G, et al. Spectrum of clinicopathologic manifestations of arrhythmogenic right
ventricular cardiomyopathy/dysplasia: a multicenter study. J Am Coll Cardiol. 1997;30:1512-20.
17. James CA, Bhonsale A, Tichnell C, et al. Exercise increases age-related penetrance and arrhythmic risk in
arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated desmosomal mutation carriers. J Am
Coll Cardiol. 2013;62:1290-7.
18. Ruwald AC, Marcus F, Estes NA 3rd, et al. Association of competitive and recreational sport participation with
cardiac events in patients with arrhythmogenic right ventricular cardiomyopathy: results from the North
American multidisciplinary study of arrhythmogenic right ventricular cardiomyopathy. Eur Heart J.
2015;36:1735-43.
19. Saberniak J, Hasselberg NE, Borgquist R, et al. Vigorous physical activity impairs myocardial function in
patients with arrhythmogenic right ventricular cardiomyopathy and in mutation positive family members. Eur J
Heart Fail. 2014;16:1337-44.
20. Sawant AC, Bhonsale A, te Riele AS, et al. Exercise has a disproportionate role in the pathogenesis of
arrhythmogenic right ventricular dysplasia/cardiomyopathy in patients without desmosomal mutations. J Am
Heart Assoc. 2014;3:e001471.
21. Sawant AC, te Riele AS, Tichnell C, et al. Safety of American Heart Association-recommended minimum
exercise for desmosomal mutation carriers. Heart Rhythm. 2016;13:199-207.
22. Bhonsale A, Groeneweg JA, James CA, et al. Impact of genotype on clinical course in arrhythmogenic right
ventricular dysplasia/cardiomyopathy-associated mutation carriers. Eur Heart J. 2015;36:847-55.
23. Bhonsale A, James CA, Tichnell C, et al. Risk stratification in arrhythmogenic right ventricular
dysplasia/cardiomyopathy-associated desmosomal mutation carriers. Circ Arrhythm Electrophysiol.
2013;6:569-78.

Page 52
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

24. Hershberger RE, Lindenfeld J, Mestroni L, et al. Genetic evaluation of cardiomyopathy--a Heart Failure Society
of America practice guideline. J Card Fail. 2009;15:83-97.
25. Kapplinger JD, Landstrom AP, Salisbury BA, et al. Distinguishing arrhythmogenic right ventricular
cardiomyopathy/dysplasia-associated mutations from background genetic noise. J Am Coll Cardiol.
2011;57:2317-27.
26. Rigato I, Bauce B, Rampazzo A, et al. Compound and digenic heterozygosity predicts lifetime arrhythmic
outcome and sudden cardiac death in desmosomal gene-related arrhythmogenic right ventricular
cardiomyopathy. Circ Cardiovasc Genet. 2013;6:533-42.
27. Philips B, te Riele AS, Sawant A, et al. Outcomes and ventricular tachycardia recurrence characteristics after
epicardial ablation of ventricular tachycardia in arrhythmogenic right ventricular dysplasia/cardiomyopathy.
Heart Rhythm. 2015;12:716-25.
28. Philips B, Madhavan S, James C, et al. Outcomes of catheter ablation of ventricular tachycardia in
arrhythmogenic right ventricular dysplasia/cardiomyopathy. Circ Arrhythm Electrophysiol. 2012;5:499-505.
29. Dalal D, Jain R, Tandri H, et al. Long-term efficacy of catheter ablation of ventricular tachycardia in patients
with arrhythmogenic right ventricular dysplasia/cardiomyopathy. J Am Coll Cardiol. 2007;50:432-40.
30. Garcia FC, Bazan V, Zado ES, et al. Epicardial substrate and outcome with epicardial ablation of ventricular
tachycardia in arrhythmogenic right ventricular cardiomyopathy/dysplasia. Circulation. 2009;120:366-75.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

31. Bai R, Di BL, Shivkumar K, et al. Ablation of ventricular arrhythmias in arrhythmogenic right ventricular
dysplasia/cardiomyopathy: arrhythmia-free survival after endo-epicardial substrate based mapping and
ablation. Circ Arrhythm Electrophysiol. 2011;4:478-85.
32. Berruezo A, Fernandez-Armenta J, Mont L, et al. Combined endocardial and epicardial catheter ablation in
arrhythmogenic right ventricular dysplasia incorporating scar dechanneling technique. Circ Arrhythm
Electrophysiol. 2012;5:111-21.
33. Santangeli P, Zado ES, Supple GE, et al. Long-term outcome with catheter ablation of ventricular tachycardia in
patients with arrhythmogenic right ventricular cardiomyopathy. Circ Arrhythm Electrophysiol. 2015;8:1413-21.
34. Choudhary N, Tompkins C, Polonsky B, et al. Clinical presentation and outcomes by sex in arrhythmogenic right
ventricular cardiomyopathy: findings from the North American ARVC registry. J Cardiovasc Electrophysiol.
2016;27:555-62.
35. Kamath GS, Zareba W, Delaney J, et al. Value of the signal-averaged electrocardiogram in arrhythmogenic right
ventricular cardiomyopathy/dysplasia. Heart Rhythm. 2011;8:256-62.
36. Saguner AM, Medeiros-Domingo A, Schwyzer MA, et al. Usefulness of inducible ventricular tachycardia to
predict long-term adverse outcomes in arrhythmogenic right ventricular cardiomyopathy. Am J Cardiol.
2013;111:250-7.

Page 53
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.4. Hypertrophic Cardiomyopathy


Recommendations for HCM
References that support the recommendations are summarized in Online Data Supplement 31.
COR LOE Recommendations
1. In patients with HCM, SCD risk stratification should be performed at the time
I B-NR
of initial evaluation and periodically thereafter (1-8).
2. In patients with HCM who have survived an SCA due to VT or VF, or have
spontaneous sustained VT causing syncope or hemodynamic compromise, an
I B-NR
ICD is recommended if meaningful survival greater than 1 year is expected (1,
6, 9, 10).
3. In first-degree relatives of patients with HCM, an ECG and echocardiogram
I B-NR
should be performed (11-17).
4. In first-degree relatives of patients with HCM due to a known causative
I B-NR mutation, genetic counseling and mutation-specific genetic testing are
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

recommended (13-15, 18, 19).


5. In patients with clinically suspected or diagnosed HCM, genetic counseling and
IIa B-NR
genetic testing are reasonable (13-15, 18-22).
6. In patients with HCM and 1 or more of the following risk factors, an ICD is
B-NR reasonable if meaningful survival of greater than 1 year is expected:
a. Maximum LV wall thickness 30 mm (LOE: B-NR) (2, 3, 23, 24).
IIa C-LD b. SCD in 1 or more first-degree relatives presumably caused by HCM (LOE:
C-LD) (25, 26).
c. 1 or more episodes of unexplained syncope within the preceding 6
C-LD months (LOE: C-LD) (8, 26).
7. In patients with HCM who have spontaneous NSVT (LOE: C-LD) (2, 26, 27) or an
B-NR abnormal blood pressure response with exercise (LOE: B-NR) (5, 28, 29), who
IIa
also have additional SCD risk modifiers or high risk features, an ICD is
C-LD reasonable if meaningful survival greater than 1 year is expected.
8. In patients with HCM who have NSVT (LOE: B-NR) (2, 26, 27) or an abnormal
B-NR
blood pressure response with exercise (LOE: B-NR) (5, 28, 29) but do not have
IIb
any other SCD risk modifiers, an ICD may be considered, but its benefit is
B-NR
uncertain.
9. In patients with HCM and a history of sustained VT or VF, amiodarone may be
IIb C-LD
considered when an ICD is not feasible or not preferred by the patient (30, 31).
III: No 10. In patients with HCM, an invasive electrophysiological study with programmed
B-NR
Benefit ventricular stimulation should not be performed for risk stratification (32, 33).
III: No 11. In patients with an identified HCM genotype in the absence of SCD risk factors,
B-NR
Benefit an ICD should not be implanted (7, 34, 35).
Table 8 and Figure7
Refer to the ACCF/AHA HCM guideline for the definition of HCM (36).

Page 54
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 8. Major Clinical Features Associated With Increased Risk of SCD in Patients With HCM
Established risk factors*
Survival from a cardiac arrest due to VT or VF (1, 5, 6)
Spontaneous sustained VT causing syncope or hemodynamic compromise (1, 5, 6)
Family history of SCD associated with HCM (25, 26)
LV wall thickness 30 mm (2, 3, 23, 24)
Unexplained syncope within 6 mo (8, 26)
NSVT 3 beats (2, 26, 27)
Abnormal blood pressure response during exercise (5, 28, 29)

Potential risk modifiers


<30 y (5, 26)
Delayed hyperenhancement on cardiac MRI (37-39, 54)
LVOT obstruction (2, 4)
Syncope >5 y ago (8, 26)
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

High-risk subsets
LV aneurysm (40, 55, 56)
LVEF <50% (52)
*There is general agreement in the literature that these factors independently convey an increased risk for SCD in
patients with HCM.
Decrease in blood pressure of 20 mm Hg or failure to increase systolic blood pressure >20 mm Hg during exertion.
There is a lack of agreement in the literature that these modifiers independently convey an increased risk of SCD in
patients with HCM; however, a risk modifier when combined with a risk factor often identifies a patient with HCM at
increased risk for SCD beyond the risk conveyed by the risk factor alone.
A small subset of patients with an LVEF <50% (end-stage disease) or an LV aneurysm warrant consideration for ICD
implantation (52).
HCM indicates hypertrophic cardiomyopathy; ICD, implantable cardioverter-defibrillator; LV, left ventricular; LVEF, left
ventricular ejection fraction; LVOT, left ventricular outflow tract; NSVT, nonsustained ventricular tachycardia; SCD,
sudden cardiac death; VT, ventricular tachycardia; and VF, ventricular fibrillation.

Page 55
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 7. Prevention of SCD in Patients With HCM


Patients with HCM

NSVT;
Family Hx SCD;
SCD survivor; abnormal BP
No LVWT >30 mm; No
sustained VT response to
syncope <6 mo
exercise

Yes
Yes No
Yes

ICD SCD
ICD
candidate* risk modifier
(Class IIa)
present
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Yes No
Yes No
ICD
ICD Amiodarone ICD ICD (Class III:
(Class I) (Class IIb) (Class IIa) (Class IIb) No Benefit)
Colors correspond to Class of Recommendation in Table 1.
See Section 7.4 in the full-text guideline for discussion.
*ICD candidacy as determined by functional status, life expectancy, or patient preference.
Risk modifiers: Age <30 y, late gadolinium enhancement on cardiac MRI, LVOT obstruction, LV aneurysm, syncope >5 y.
BP indicates blood pressure; HCM, hypertrophic cardiomyopathy; Hx, history; ICD, implantable cardioverter-defibrillator;
LVOT, left ventricular outflow tract; LVWT, left ventricular wall thickness; MRI, magnetic resonance imaging; NSVT,
nonsustained ventricular tachycardia; SCD, sudden cardiac death; and VT, ventricular tachycardia.

References
1. Elliott PM, Sharma S, Varnava A, et al. Survival after cardiac arrest or sustained ventricular tachycardia in patients
with hypertrophic cardiomyopathy. J Am Coll Cardiol. 1999;33:1596-601.
2. Elliott PM, Poloniecki J, Dickie S, et al. Sudden death in hypertrophic cardiomyopathy: identification of high risk
patients. J Am Coll Cardiol. 2000;36:2212-8.
3. Elliott PM, Gimeno Blanes JR, Mahon NG, et al. Relation between severity of left-ventricular hypertrophy and
prognosis in patients with hypertrophic cardiomyopathy. Lancet. 2001;357:420-4.
4. Elliott PM, Gimeno JR, Tome MT, et al. Left ventricular outflow tract obstruction and sudden death risk in patients
with hypertrophic cardiomyopathy. Eur Heart J. 2006;27:1933-41.
5. Maki S, Ikeda H, Muro A, et al. Predictors of sudden cardiac death in hypertrophic cardiomyopathy. Am J Cardiol.
1998;82:774-8.
6. Maron BJ, Spirito P, Shen WK, et al. Implantable cardioverter-defibrillators and prevention of sudden cardiac death
in hypertrophic cardiomyopathy. JAMA. 2007;298:405-12.
7. O'Mahony C, Jichi F, Pavlou M, et al. A novel clinical risk prediction model for sudden cardiac death in hypertrophic
cardiomyopathy (HCM risk-SCD). Eur Heart J. 2014;35:2010-20.
8. Spirito P, Autore C, Rapezzi C, et al. Syncope and risk of sudden death in hypertrophic cardiomyopathy. Circulation.
2009;119:1703-10.
9. O'Mahony C, Lambiase PD, Quarta G, et al. The long-term survival and the risks and benefits of implantable
cardioverter defibrillators in patients with hypertrophic cardiomyopathy. Heart. 2012;98:116-25.
10.Syska P, Przybylski A, Chojnowska L, et al. Implantable cardioverter-defibrillator in patients with hypertrophic
cardiomyopathy: efficacy and complications of the therapy in long-term follow-up. J Cardiovasc Electrophysiol.
2010;21:883-9.
11.Adabag AS, Kuskowski MA, Maron BJ. Determinants for clinical diagnosis of hypertrophic cardiomyopathy. Am J
Cardiol. 2006;98:1507-11.

Page 56
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

12.Afonso LC, Bernal J, Bax JJ, et al. Echocardiography in hypertrophic cardiomyopathy: the role of conventional and
emerging technologies. JACC Cardiovasc Imag. 2008;1:787-800.
13.Girolami F, Ho CY, Semsarian C, et al. Clinical features and outcome of hypertrophic cardiomyopathy associated with
triple sarcomere protein gene mutations. J Am Coll Cardiol. 2010;55:1444-53.
14.Ingles J, Sarina T, Yeates L, et al. Clinical predictors of genetic testing outcomes in hypertrophic cardiomyopathy.
Genet Med. 2013;15:972-7.
15.Jensen MK, Havndrup O, Christiansen M, et al. Penetrance of hypertrophic cardiomyopathy in children and
adolescents: a 12-year follow-up study of clinical screening and predictive genetic testing. Circulation. 2013;127:48-
54.
16.Klues HG, Schiffers A, Maron BJ. Phenotypic spectrum and patterns of left ventricular hypertrophy in hypertrophic
cardiomyopathy: morphologic observations and significance as assessed by two-dimensional echocardiography in
600 patients. J Am Coll Cardiol. 1995;26:1699-708.
17.Wigle ED, Rakowski H, Kimball BP, et al. Hypertrophic cardiomyopathy. Clinical spectrum and treatment. Circulation.
1995;92:1680-92.
18.Christiaans I, Birnie E, van Langen IM, et al. The yield of risk stratification for sudden cardiac death in hypertrophic
cardiomyopathy myosin-binding protein C gene mutation carriers: focus on predictive screening. Eur Heart J.
2010;31:842-8.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

19.Olivotto I, Girolami F, Ackerman MJ, et al. Myofilament protein gene mutation screening and outcome of patients
with hypertrophic cardiomyopathy. Mayo Clin Proc. 2008;83:630-8.
20.Christiaans I, van Langen IM, Birnie E, et al. Genetic counseling and cardiac care in predictively tested hypertrophic
cardiomyopathy mutation carriers: the patients' perspective. Am J Med Genet. 2009;149a:1444-51.
21.Hamang A, Eide GE, Rokne B, et al. Predictors of heart-focused anxiety in patients undergoing genetic investigation
and counseling of long QT syndrome or hypertrophic cardiomyopathy: a one year follow-up. J Genet Counsel.
2012;21:72-84.
22.Bos JM, Will ML, Gersh BJ, et al. Characterization of a phenotype-based genetic test prediction score for unrelated
patients with hypertrophic cardiomyopathy. Mayo Clin Proc. 2014;89:727-37.
23.Sorajja P, Nishimura RA, Ommen SR, et al. Use of echocardiography in patients with hypertrophic cardiomyopathy:
clinical implications of massive hypertrophy. J Am Soc Echocardiogr. 2006;19:788-95.
24.Spirito P, Bellone P, Harris KM, et al. Magnitude of left ventricular hypertrophy and risk of sudden death in
hypertrophic cardiomyopathy. N Engl J Med. 2000;342:1778-85.
25.Bos JM, Maron BJ, Ackerman MJ, et al. Role of family history of sudden death in risk stratification and prevention of
sudden death with implantable defibrillators in hypertrophic cardiomyopathy. Am J Cardiol. 2010;106:1481-6.
26.Maron BJ, Shen WK, Link MS, et al. Efficacy of implantable cardioverter-defibrillators for the prevention of sudden
death in patients with hypertrophic cardiomyopathy. N Engl J Med. 2000;342:365-73.
27.Monserrat L, Elliott PM, Gimeno JR, et al. Non-sustained ventricular tachycardia in hypertrophic cardiomyopathy: an
independent marker of sudden death risk in young patients. J Am Coll Cardiol. 2003;42:873-9.
28.Olivotto I, Maron BJ, Montereggi A, et al. Prognostic value of systemic blood pressure response during exercise in a
community-based patient population with hypertrophic cardiomyopathy. J Am Coll Cardiol. 1999;33:2044-51.
29.Sadoul N, Prasad K, Elliott PM, et al. Prospective prognostic assessment of blood pressure response during exercise
in patients with hypertrophic cardiomyopathy. Circulation. 1997;96:2987-91.
30.McKenna WJ, Oakley CM, Krikler DM, et al. Improved survival with amiodarone in patients with hypertrophic
cardiomyopathy and ventricular tachycardia. Br Heart J. 1985;53:412-6.
31.Melacini P, Maron BJ, Bobbo F, et al. Evidence that pharmacological strategies lack efficacy for the prevention of
sudden death in hypertrophic cardiomyopathy. Heart. 2007;93:708-10.
32.Kuck KH, Kunze KP, Schluter M, et al. Programmed electrical stimulation in hypertrophic cardiomyopathy. Results in
patients with and without cardiac arrest or syncope. Eur Heart J. 1988;9:177-85.
33.Zhu DW, Sun H, Hill R, et al. The value of electrophysiology study and prophylactic implantation of cardioverter
defibrillator in patients with hypertrophic cardiomyopathy. Pacing Clin Electrophysiol. 1998;21:299-302.
34.Ackerman MJ, VanDriest SL, Ommen SR, et al. Prevalence and age-dependence of malignant mutations in the beta-
myosin heavy chain and troponin T genes in hypertrophic cardiomyopathy: a comprehensive outpatient perspective.
J Am Coll Cardiol. 2002;39:2042-8.
35.Lopes LR, Rahman MS, Elliott PM. A systematic review and meta-analysis of genotype-phenotype associations in
patients with hypertrophic cardiomyopathy caused by sarcomeric protein mutations. Heart. 2013;99:1800-11.

Page 57
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.5. Myocarditis
Recommendations for Myocarditis
References that support the recommendations are summarized in Online Data Supplement 32.
COR LOE Recommendations
1. In patients with life-threatening VT or VF associated with confirmed or clinically
I C-LD suspected myocarditis, referral to centers with mechanical hemodynamic
support and advanced arrhythmia management is recommended (1).
2. In patients with giant cell myocarditis with VF or hemodynamically unstable VT
IIb C-LD treated according to GDMT, an ICD and/or an antiarrhythmic medication may be
considered if meaningful survival of greater than 1 year is expected (2-4).

References
1. Aoyama N, Izumi T, Hiramori K, et al. National survey of fulminant myocarditis in Japan: therapeutic guidelines and
long-term prognosis of using percutaneous cardiopulmonary support for fulminant myocarditis (special report from
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

a scientific committee). Circ J. 2002;66:133-44.


2. Maleszewski JJ, Orellana VM, Hodge DO, et al. Long-term risk of recurrence, morbidity and mortality in giant cell
myocarditis. Am J Cardiol. 2015;115:1733-8.
3. Cooper LT Jr, Berry GJ, Shabetai R. Idiopathic giant-cell myocarditis--natural history and treatment. Multicenter
Giant Cell Myocarditis Study Group Investigators. N Engl J Med. 1997;336:1860-6.
4. Kandolin R, Lehtonen J, Salmenkivi K, et al. Diagnosis, treatment, and outcome of giant-cell myocarditis in the era of
combined immunosuppression. Circ Heart Fail. 2013;6:15-22.

6.6. Cardiac Sarcoidosis


Recommendations for Cardiac Sarcoidosis
References that support the recommendations are summarized in Online Data Supplement 33.
COR LOE Recommendations
1. In patients with cardiac sarcoidosis who have sustained VT or are survivors of
I B-NR SCA or have an LVEF of 35% or less, an ICD is recommended, if meaningful
survival of greater than 1 year is expected (1-5).
2. In patients with cardiac sarcoidosis and LVEF greater than 35% who have
syncope and/or evidence of myocardial scar by cardiac MRI or positron
IIa B-NR emission tomographic (PET) scan, and/or have an indication for permanent
pacing implantation of an ICD is reasonable, provided that meaningful survival
of greater than 1 year is expected (6-10).
3. In patients with cardiac sarcoidosis and LVEF greater than 35%, it is reasonable
to perform an electrophysiological study and to impant an ICD, if sustained VA
IIa C-LD
is inducible, provided that meaningful survival of greater than 1 year is
expected (11, 12).
4. In patients with cardiac sarcoidosis who have an indication for permanent
IIa C-LD
pacing, implantation of an ICD can be beneficial (13).
5. In patients with cardiac sarcoidosis with frequent symptomatic VA and
evidence of myocardial inflammation, immunosuppression in combination
IIa C-LD
with antiarrhythmic medication therapy can be useful to reduce VA burden
(14-16).
Figure 8

Page 58
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 8. Prevention of SCD in Patients With Cardiac Sarcoidosis

Syncope, and/or
SCA survivor/
extensive scar by
Patients with cardiac sustained VT, LVEF
No No cardiac MRI or PET
sarcoidosis either spontaneous 35%
and/or a candidate for
or inducible
permanent pacemaker

Yes
Yes
Yes

ICD ICD
candidate* candidate*
ICD
candidate*

Yes No Yes No
Yes No
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

ICD GDMT ICD GDMT


(Class I) (Class I) (Class I) (Class I) EP study GDMT
or ICD (Class I)
(Class IIa)

Recurrent
Immunosuppression
VA and Yes
(Class IIa)
Inflammation

Colors correspond to Class of Recommendation in Table 1.


See Section 7.6 in the full-text guideline for discussion.
*ICD candidacy as determined by functional status, life expectancy, or patient preference.
For recurrent sustained monomorphic VT, refer to Figure 2.
CEP indicates electrophysiological; GDMT, guideline-directed management and therapy; ICD, implantable cardiac-
defibrillator; LVEF, left ventricular ejection fraction; MRI, magnetic resonance imaging; PET, positron emission
tomography; SCA, sudden cardiac arrest; SCD, sudden cardiac death; VA, ventricular arrhythmia; and VT, ventricular
tachycardia.

References
1. Kandolin R, Lehtonen J, Airaksinen J, et al. Cardiac sarcoidosis: epidemiology, characteristics, and outcome over 25
years in a nationwide study. Circulation. 2015;131:624-32.
2. Yazaki Y, Isobe M, Hiroe M, et al. Prognostic determinants of long-term survival in Japanese patients with cardiac
sarcoidosis treated with prednisone. Am J Cardiol. 2001;88:1006-10.
3. Kron J, Sauer W, Schuller J, et al. Efficacy and safety of implantable cardiac defibrillators for treatment of ventricular
arrhythmias in patients with cardiac sarcoidosis. Europace. 2013;15:347-54.
4. Mohsen A, Jimenez A, Hood RE, et al. Cardiac sarcoidosis: electrophysiological outcomes on long-term follow-up and
the role of the implantable cardioverter-defibrillator. J Cardiovasc Electrophysiol. 2014;25:171-6.
5. Schuller JL, Zipse M, Crawford T, et al. Implantable cardioverter defibrillator therapy in patients with cardiac
sarcoidosis. J Cardiovasc Electrophysiol. 2012;23:925-9.
6. Coleman GC, Shaw PW, Balfour PC Jr, et al. Prognostic value of myocardial scarring on CMR in patients with cardiac
sarcoidosis: a systematic review and meta-analysis. JACC Cardiovasc Imaging. 2016;10:411-20.
7. Murtagh G, Laffin LJ, Beshai JF, et al. Prognosis of myocardial damage in sarcoidosis patients with preserved left
ventricular ejection fraction: risk stratification using cardiovascular magnetic resonance. Circ Cardiovasc Imaging.
2016;9:e003738.
8. Crawford T, Mueller G, Sarsam S, et al. Magnetic resonance imaging for identifying patients with cardiac sarcoidosis
and preserved or mildly reduced left ventricular function at risk of ventricular arrhythmias. Circ Arrhythm
Electrophysiol. 2014;7:1109-15.

Page 59
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

9. Greulich S, Deluigi CC, Gloekler S, et al. CMR imaging predicts death and other adverse events in suspected cardiac
sarcoidosis. JACC Cardiovasc Imaging. 2013;6:501-11.
10.Blankstein R, Osborne M, Naya M, et al. Cardiac positron emission tomography enhances prognostic assessments of
patients with suspected cardiac sarcoidosis. J Am Coll Cardiol. 2014;63:329-36.
11.Aizer A, Stern EH, Gomes JA, et al. Usefulness of programmed ventricular stimulation in predicting future arrhythmic
events in patients with cardiac sarcoidosis. Am J Cardiol. 2005;96:276-82.
12.Mehta RH, Starr AZ, Lopes RD, et al. Incidence of and outcomes associated with ventricular tachycardia or fibrillation
in patients undergoing primary percutaneous coronary intervention. JAMA. 2009;301:1779-89.
13.Takaya Y, Kusano KF, Nakamura K, et al. Outcomes in patients with high-degree atrioventricular block as the initial
manifestation of cardiac sarcoidosis. Am J Cardiol 2015;115:505-9.
14.Naruse Y, Sekiguchi Y, Nogami A, et al. Systematic treatment approach to ventricular tachycardia in cardiac
sarcoidosis. Circ Arrhythm Electrophysiol. 2014;7:407-13.
15.Segawa M, Fukuda K, Nakano M, et al. Time course and factors correlating with ventricular tachyarrhythmias after
introduction of steroid therapy in cardiac sarcoidosis. Circ Arrhythm Electrophysiol. 2016;9:e003353.
16.Yodogawa K, Seino Y, Ohara T, et al. Effect of corticosteroid therapy on ventricular arrhythmias in patients with
cardiac sarcoidosis. Ann Noninvasive Electrocardiol. 2011;16:140-7.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

6.7. Heart Failure


6.7.1. HF With Reduced Ejection Fraction
Recommendation for HFrEF
References that support the recommendation are summarized in Online Data Supplement 35.
COR LOE Recommendation
1. In patients with HFrEF who are awaiting heart transplant and who otherwise
IIa B-NR would not qualify for an ICD (e.g., NYHA class IV and/or use of inotropes) with
a plan to discharge home, an ICD is reasonable (1-5).

References
1. Frohlich GM, Holzmeister J, Hubler M, et al. Prophylactic implantable cardioverter defibrillator treatment in patients
with end-stage heart failure awaiting heart transplantation. Heart. 2013;99:1158-65.
2. Kao AC, Krause SW, Handa R, et al. Wearable defibrillator use in heart failure (WIF): results of a prospective registry.
BMC Cardiovasc Disord. 2012;12:123.
3. Opreanu M, Wan C, Singh V, et al. Wearable cardioverter-defibrillator as a bridge to cardiac transplantation: a
national database analysis. J Heart Lung Transplant. 2015;34:1305-9.
4. Sandner SE, Wieselthaler G, Zuckermann A, et al. Survival benefit of the implantable cardioverter-defibrillator in
patients on the waiting list for cardiac transplantation. Circulation. 2001;104:I171-6.
5. Vakil K, Duval S, Cogswell R, et al. Impact of implantable cardioverter-defibrillators on waitlist mortality
among patients awaiting heart transplantation. An UNOS/OPTN Analysis. JACC Clin Electrophysiol. 2017;3:33-40.

6.7.2. Left Ventricular Assist Device


Recommendation for Patients With an LVAD
References that support the recommendation are summarized in Online Data Supplement 36.
COR LOE Recommendation
IIa C-LD 1. In patients with an LVAD and sustained VA, an ICD can be beneficial (1).

References
1. Vakil K, Kazmirczak F, Sathnur N, et al. Implantable cardioverter-defibrillator use in patients with left ventricular
assist devices: a systematic review and meta-analysis. JACC Heart Fail. 2016;4:772-9.

Page 60
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.7.3. ICD Use After Heart Transplantation


Recommendation for ICD Use After Heart Transplantation
References that support the recommendation are summarized in Online Data Supplement 37.
COR LOE Recommendation
1. In patients with a heart transplant and severe allograft vasculopathy with LV
IIb B-NR dysfunction, an ICD may be reasonable if meaningful survival of greater than
1 year is expected (1-3).

References
1. Tsai VW, Cooper J, Garan H, et al. The efficacy of implantable cardioverter-defibrillators in heart transplant
recipients: results from a multicenter registry. Circ Heart Fail. 2009;2:197-201.
2. McDowell DL, Hauptman PJ. Implantable defibrillators and cardiac resynchronization therapy in heart transplant
recipients: results of a national survey. J Heart Lung Transplant. 2009;28:847-50.
3. Neylon A, Canniffe C, Parlon B, et al. Implantable cardioverter-defibrillators in a heart transplant population: a
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

single-center experience. J Heart Lung Transplant. 2016;35:682-4.

6.8. Neuromuscular Disorders


Recommendations for Neuromuscular Disorders
References that support the recommendations are summarized in Online Data Supplement 38.
COR LOE Recommendations
1. In patients with neuromuscular disorders, primary and secondary prevention
I B-NR ICDs are recommended for the same indications as for patients with NICM if
meaningful survival of greater than 1 year is expected (1, 2).
2. In patients with Emery-Dreifuss and limb-girdle type IB muscular dystrophies
IIa B-NR with progressive cardiac involvement, an ICD is reasonable if a meaningful
survival of greater than 1 year is expected (3-8).
3. In patients with muscular dystrophy, follow-up for development of cardiac
IIa B-NR involvement is reasonable, even if the patient is asymptomatic at presentation
(9-12).
4. In patients with myotonic dystrophy type 1 with an indication for a permanent
IIb B-NR pacemaker, an ICD may be considered to minimize the risk of SCA from VT if
meaningful survival of greater than 1 year is expected (9, 13, 14).
Table 9

Page 61
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 9. Neuromuscular Disorders Associated With Heart Disease


Gene/ Primary Frequency
Muscular Protein Cardiac of Cardiac Associated With
Dystrophy Inheritance Affected Pathology Involvement Causes of Death Sudden Death?
Duchenne X-linked Dystrophin NICM >90% Respiratory, HF Yes, uncertain
recessive etiology
Becker X-linked Dystrophin NICM 60%75% HF, respiratory Yes, uncertain
recessive etiology
Limb-girdle type Autosomal Lamin A/C Conduction >90% Sudden, HF Yes
1B dominant system
disease and
NICM
Limb-girdle type Autosomal Sarcoglycan NICM <25% Respiratory, HF Uncertain
2C-2F recessive
Limb-girdle type Autosomal Fukutin- NICM 20%80% Respiratory, HF Uncertain
2I recessive related
protein
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Myotonic type 1 Autosomal CTG repeat Conduction 60%80% Respiratory, 30% of deaths,
dominant expansion system sudden, HF uncertain
disease and bradycardia
NICM versus
tachycardia
Myotonic type 2 Autosomal CCTG repeat Conduction 10%25% Normal causes Reported
dominant expansion system
disease
Emery-Dreifuss X-linked and Emerin, Conduction >90% Sudden, HF Yes
autosomal Lamin A/C system
dominant or disease and
recessive NICM
Facioscapulohu Autosomal D4Z4 repeat Possibly 5%15% Normal causes, Not reported
meral dominant contaction conduction respiratory rarely
disease
HF indicates heart failure; and NICM, nonischemic cardiomyopathy.
Adapted with permission from Groh, et al. (15).

References
1. Bhakta D, Groh MR, Shen C, et al. Increased mortality with left ventricular systolic dysfunction and heart failure in
adults with myotonic dystrophy type 1. Am Heart J. 2010;160:1137-41, 41.
2. Merino JL, Carmona JR, Fernandez-Lozano I, et al. Mechanisms of sustained ventricular tachycardia in myotonic
dystrophy: implications for catheter ablation. Circulation. 1998;98:541-6.
3. Anselme F, Moubarak G, Savoure A, et al. Implantable cardioverter-defibrillators in lamin A/C mutation carriers with
cardiac conduction disorders. Heart Rhythm. 2013;10:1492-8.
4. van Rijsingen IA, Arbustini E, Elliott PM, et al. Risk factors for malignant ventricular arrhythmias in lamin A/C
mutation carriers a European cohort study. J Am Coll Cardiol. 2012;59:493-500.
5. Meune C, Van Berlo JH, Anselme F, et al. Primary prevention of sudden death in patients with lamin A/C gene
mutations. N Engl J Med. 2006;354:209-10.
6. Pasotti M, Klersy C, Pilotto A, et al. Long-term outcome and risk stratification in dilated cardiolaminopathies. J Am
Coll Cardiol. 2008;52:1250-60.
7. Van Berlo JH, de Voogt WG, van der Kooi AJ, et al. Meta-analysis of clinical characteristics of 299 carriers of LMNA
gene mutations: do lamin A/C mutations portend a high risk of sudden death? J Mol Med. 2005;83:79-83.
8. Russo V, Nigro G. ICD role in preventing sudden cardiac death in Emery-Dreifuss muscular dystrophy with preserved
myocardial function: 2013 ESC Guidelines on Cardiac Pacing and Cardiac Resynchronization Therapy. Europace.
2015;17:337.

Page 62
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

9. Groh WJ, Groh MR, Saha C, et al. Electrocardiographic abnormalities and sudden death in myotonic dystrophy type
1. N Engl J Med. 2008;358:2688-97.
10.Lallemand B, Clementy N, Bernard-Brunet A, et al. The evolution of infrahissian conduction time in myotonic
dystrophy patients: clinical implications. Heart. 2012;98:291-6.
11.Nazarian S, Wagner KR, Caffo BS, et al. Clinical predictors of conduction disease progression in type I myotonic
muscular dystrophy. Pacing Clin Electrophysiol. 2011;34:171-6.
12.Tanawuttiwat T, Wagner KR, Tomaselli G, et al. Left ventricular dysfunction and conduction disturbances in patients
with myotonic muscular dystrophy type I and II. JAMA Cardiology. 2017;2:225-8.
13.Bhakta D, Shen C, Kron J, et al. Pacemaker and implantable cardioverter-defibrillator use in a US myotonic dystrophy
type 1 population. J Cardiovasc Electrophysiol. 2011;22:1369-75.
14.Laurent V, Pellieux S, Corcia P, et al. Mortality in myotonic dystrophy patients in the area of prophylactic pacing
devices. Int J Cardiol. 2011;150:54-8.
15.Groh WJ. Arrhythmias in the muscular dystrophies. Heart Rhythm. 2012;9:1890-95.

6.9. Cardiac Channelopathies


Recommendations for Cardiac Channelopathies
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

References that support the recommendations are summarized in Online Data Supplement 39.
COR LOE Recommendations
1. In first-degree relatives of patients who have a causative mutation for long QT
syndrome, catecholaminergic polymorphic ventricular tachycardia, short QT
I B-NR
syndrome, or Brugada syndrome, genetic counseling and mutation-specific
genetic testing are recommended (1-6).
2. In patients with a cardiac channelopathy and SCA, an ICD is recommended if
I B-NR
meaningful survival of greater than 1 year is expected (7-13).

References
1. Bai R, Napolitano C, Bloise R, et al. Yield of genetic screening in inherited cardiac channelopathies: how to prioritize
access to genetic testing. Circ Arrhythm Electrophysiol. 2009;2:6-15.
2. Goldenberg I, Horr S, Moss AJ, et al. Risk for life-threatening cardiac events in patients with genotype-confirmed
long-QT syndrome and normal-range corrected QT intervals. J Am Coll Cardiol. 2011;57:51-9.
3. Nannenberg EA, Sijbrands EJ, Dijksman LM, et al. Mortality of inherited arrhythmia syndromes: insight into their
natural history. Circ Cardiovasc Genet. 2012;5:183-9.
4. Priori SG, Gasparini M, Napolitano C, et al. Risk stratification in Brugada syndrome: results of the PRELUDE
(PRogrammed ELectrical stimUlation preDictive valuE) registry. J Am Coll Cardiol. 2012;59:37-45.
5. Priori SG, Napolitano C, Memmi M, et al. Clinical and molecular characterization of patients with catecholaminergic
polymorphic ventricular tachycardia. Circulation. 2002;106:69-74.
6. Wilde AA, Moss AJ, Kaufman ES, et al. Clinical aspects of type 3 long-QT syndrome: an international multicenter
study. Circulation. 2016;134:872-82.
7. Wedekind H, Burde D, Zumhagen S, et al. QT interval prolongation and risk for cardiac events in genotyped LQTS-
index children. Eur J Pediatr. 2009;168:1107-15.
8. Moss AJ, Zareba W, Hall WJ, et al. Effectiveness and limitations of beta-blocker therapy in congenital long-QT
syndrome. Circulation. 2000;101:616-23.
9. Zareba W, Moss AJ, Daubert JP, et al. Implantable cardioverter defibrillator in high-risk long QT syndrome patients. J
Cardiovasc Electrophysiol. 2003;14:337-41.
10.Monnig G, Kobe J, Loher A, et al. Implantable cardioverter-defibrillator therapy in patients with congenital long-QT
syndrome: a long-term follow-up. Heart Rhythm. 2005;2:497-504.
11.Hayashi M, Denjoy I, Extramiana F, et al. Incidence and risk factors of arrhythmic events in catecholaminergic
polymorphic ventricular tachycardia. Circulation. 2009;119:2426-34.
12.Gehi AK, Duong TD, Metz LD, et al. Risk stratification of individuals with the Brugada electrocardiogram: a meta-
analysis. J Cardiovasc Electrophysiol. 2006;17:577-83.
13.Probst V, Veltmann C, Eckardt L, et al. Long-term prognosis of patients diagnosed with Brugada syndrome: results
from the FINGER Brugada syndrome registry. Circulation. 2010;121:635-43.

Page 63
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.9.1. Specific Cardiac Channelopathy Syndromes


6.9.1.1. Congenital Long QT Syndrome
Recommendations for Long QT Syndrome
References that support the recommendations are summarized in Online Data Supplement 40.
COR LOE Recommendations
1. In patients with long QT syndrome with a resting QTc greater than 470 ms, a
I B-NR
beta blocker is recommended (1-5).
2. In high-risk patients with symptomatic long QT syndrome in whom a beta
blocker is ineffective or not tolerated, intensification of therapy with additional
I B-NR
medications (guided by consideration of the particular long QT syndrome type),
left cardiac sympathetic denervation, and/or an ICD is recommended (2, 6-12).
3. In patients with long QT syndrome and recurrent appropriate ICD shocks
despite maximum tolerated doses of a beta blocker, intensification of medical
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

I B-NR therapy with additional medications (guided by consideration of according to


the particular long QT syndrome type) or left cardiac sympathetic denervation,
is recommended (6, 7, 10, 13-16).
4. In patients with clinically diagnosed long QT syndrome, genetic counseling and
I B-NR
genetic testing are recommended (17-21).
5. In patients with suspected long QT syndrome, ambulatory electrocardiographic
monitoring, recording the ECG lying and immediately on standing, and/or
IIa B-NR
exercise treadmill testing can be useful for establishing a diagnosis and
monitoring the response to therapy (22-29).
6. In asymptomatic patients with long QT syndrome and a resting QTc less than
IIa B-NR
470 ms, chronic therapy with a beta blocker is reasonable (3, 30, 31).
7. In asymptomatic patients with long QT syndrome and a resting QTc greater
than 500 ms while receiving a beta blocker, intensification of therapy with
IIb B-NR
medications (guided by consideration of the particular long QT syndrome type),
left cardiac sympathetic denervation or an ICD may be considered (2, 8, 11, 30).
III: 8. In patients with long QT syndrome, QT-prolonging medications are potentially
B-NR
Harm harmful (5, 12, 32-34).
Table 10 and Figures 9, 10 (LQT1), 11 (LQT2), and 12 (LQT3)

Page 64
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 10. Commonly Used QT-Prolonging Medications (35,36)


Examples of QT Prolonging Medications*
Antiarrhythmic Medications Psychotropic Medications Antibiotics Others
Disopyramide Haloperidol Erythromycin Methadone
Procainamide (N- Phenothiazines Pentamidine Probucol
acetylprocainamide) Citalopram Azithromycin Droperidol
Quinidine Tricyclic antidepressants Chloroquine Ondansetron
Dofetilide Ciprofloxacin
Dronedarone Fluconazole
Ibutilide Levofloxacin
Sotalol Moxifloxacin
Amiodarone Clarithromycin
Itraconazole
Ketoconazole
*A more complete list is maintained at: www.crediblemeds.org (35).
Amiodarone rarely causes torsades de pointes.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Figure 9. Prevention of SCD in Patients With Long QT Syndrome

LQTS QT prolonging drugs/


hypokalemia/
Resuscitated hypomagnesemia
cardiac arrest (Class III: Harm)
QTc <470 ms QTc 470 ms and/
or symptomatic
ICD Beta blocker
candidate* (Class I)
Beta blocker Beta blocker
(Class IIa) (Class I)
ICD
(Class I)
Persistent symptoms
Recurrent ICD Asymptomatic and
and/or other high-risk
shocks for VT QTc >500 ms
features

Treatment intensification: Treatment intensification: Treatment intensification:


additional medications, additional medications, additional medications,
left cardiac sympathetic left cardiac sympathetic left cardiac sympathetic
denervation denervation and/or an ICD denervation and/or an ICD
(Class I) (Class I) (Class IIb)
Colors correspond to Class of Recommendation in Table 1.
See Section 7.9.1.1 in the full-text guideline for discussion.
*ICD candidacy as determined by functional status, life expectancy, or patient preference.
High-risk patients with LQTS include those with QTc >500 ms, genotypes LQT2 and LQT3, females with genotype LQT2,
<40 years of age, onset of symptoms at <10 years of age, and patients with recurrent syncope.
ICD indicates implantable cardioverter-defibrillator; LQTS, long-QT syndrome; VT, ventricular tachycardia.

Page 65
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 10. Long-QT Syndrome Type 1

Figure 11. Long-QT Syndrome Type 2


Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Figure 12. Long-QT Syndrome Type 3

References
1. Abu-Zeitone A, Peterson DR, Polonsky B, et al. Efficacy of different beta-blockers in the treatment of long QT
syndrome. J Am Coll Cardiol. 2014;64:1352-8.
2. Goldenberg I, Bradley J, Moss A, et al. Beta-blocker efficacy in high-risk patients with the congenital long-QT
syndrome types 1 and 2: implications for patient management. J Cardiovasc Electrophysiol. 2010;21:893-901.
3. Moss AJ, Zareba W, Hall WJ, et al. Effectiveness and limitations of beta-blocker therapy in congenital long-QT
syndrome. Circulation. 2000;101:616-23.
4. Sauer AJ, Moss AJ, McNitt S, et al. Long QT syndrome in adults. J Am Coll Cardiol. 2007;49:329-37.
5. Vincent GM, Schwartz PJ, Denjoy I, et al. High efficacy of beta-blockers in long-QT syndrome type 1: contribution of
noncompliance and QT-prolonging drugs to the occurrence of beta-blocker treatment "failures". Circulation.
2009;119:215-21.
6. Chorin E, Hu D, Antzelevitch C, et al. Ranolazine for congenital long-qt syndrome type iii: experimental and long-
term clinical data. Circ Arrhythm Electrophysiol. 2016;9:e004370.

Page 66
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

7. Chorin E, Taub R, Medina A, et al. Long-term flecainide therapy in type 3 long QT syndrome. Europace. 2017;euw439
[Epub ahead of print].
8. Hobbs JB, Peterson DR, Moss AJ, et al. Risk of aborted cardiac arrest or sudden cardiac death during adolescence in
the long-QT syndrome. JAMA. 2006;296:1249-54.
9. Jons C, Moss AJ, Goldenberg I, et al. Risk of fatal arrhythmic events in long QT syndrome patients after syncope. J
Am Coll Cardiol. 2010;55:783-8.
10.Mazzanti A, Maragna R, Faragli A, et al. Gene-specific therapy with mexiletine reduces arrhythmic events in patients
with long QT syndrome type 3. J Am Coll Cardiol. 2016;67:1053-8.
11.Nannenberg EA, Sijbrands EJ, Dijksman LM, et al. Mortality of inherited arrhythmia syndromes: insight into their
natural history. Circ Cardiovasc Genet. 2012;5:183-9.
12.Wedekind H, Burde D, Zumhagen S, et al. QT interval prolongation and risk for cardiac events in genotyped LQTS-
index children. Eur J Pediatr. 2009;168:1107-15.
13.Collura CA, Johnson JN, Moir C, et al. Left cardiac sympathetic denervation for the treatment of long QT syndrome
and catecholaminergic polymorphic ventricular tachycardia using video-assisted thoracic surgery. Heart Rhythm.
2009;6:752-9.
14.Hofferberth SC, Cecchin F, Loberman D, et al. Left thoracoscopic sympathectomy for cardiac denervation in patients
with life-threatening ventricular arrhythmias. J Thorac Cardiovasc Surg. 2014;147:404-9.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

15.Schneider HE, Steinmetz M, Krause U, et al. Left cardiac sympathetic denervation for the management of life-
threatening ventricular tachyarrhythmias in young patients with catecholaminergic polymorphic ventricular
tachycardia and long QT syndrome. Clin Res Cardiol. 2013;102:33-42.
16.Schwartz PJ, Priori SG, Cerrone M, et al. Left cardiac sympathetic denervation in the management of high-risk
patients affected by the long-QT syndrome. Circulation. 2004;109:1826-33.
17.Bai R, Napolitano C, Bloise R, et al. Yield of genetic screening in inherited cardiac channelopathies: how to prioritize
access to genetic testing. Circ Arrhythm Electrophysiol. 2009;2:6-15.
18.Costa J, Lopes CM, Barsheshet A, et al. Combined assessment of sex- and mutation-specific information for risk
stratification in type 1 long QT syndrome. Heart Rhythm. 2012;9:892-8.
19.Kim JA, Lopes CM, Moss AJ, et al. Trigger-specific risk factors and response to therapy in long QT syndrome type 2.
Heart Rhythm. 2010;7:1797-805.
20.Migdalovich D, Moss AJ, Lopes CM, et al. Mutation and gender-specific risk in type 2 long QT syndrome: implications
for risk stratification for life-threatening cardiac events in patients with long QT syndrome. Heart Rhythm.
2011;8:1537-43.
21.Tester DJ, Will ML, Haglund CM, et al. Effect of clinical phenotype on yield of long QT syndrome genetic testing. J Am
Coll Cardiol. 2006;47:764-8.
22.Adler A, van der Werf C, Postema PG, et al. The phenomenon of QT stunning: the abnormal QT prolongation
provoked by standing persists even as the heart rate returns to normal in patients with long QT syndrome. Heart
Rhythm. 2012;9:901-8.
23.Aziz PF, Wieand TS, Ganley J, et al. Genotype- and mutation site-specific QT adaptation during exercise, recovery,
and postural changes in children with long-QT syndrome. Circ Arrhythm Electrophysiol. 2011;4:867-73.
24.Chattha IS, Sy RW, Yee R, et al. Utility of the recovery electrocardiogram after exercise: a novel indicator for the
diagnosis and genotyping of long QT syndrome? Heart Rhythm. 2010;7:906-11.
25.Laksman ZW, Hamilton RM, Chockalingam P, et al. Mutation location effect on severity of phenotype during exercise
testing in type 1 long-QT syndrome: impact of transmembrane and C-loop location. J Cardiovasc Electrophysiol.
2013;24:1015-20.
26.Moltedo JM, Kim JJ, Friedman RA, et al. Use of a cardioselective beta-blocker for pediatric patients with prolonged
QT syndrome. Pediatr Cardiol. 2011;32:63-6.
27.Sy RW, van der Werf C, Chattha IS, et al. Derivation and validation of a simple exercise-based algorithm for
prediction of genetic testing in relatives of LQTS probands. Circulation. 2011;124:2187-94.
28.Villain E, Denjoy I, Lupoglazoff JM, et al. Low incidence of cardiac events with beta-blocking therapy in children with
long QT syndrome. Eur Heart J. 2004;25:1405-11.
29.Viskin S, Postema PG, Bhuiyan ZA, et al. The response of the QT interval to the brief tachycardia provoked by
standing: a bedside test for diagnosing long QT syndrome. J Am Coll Cardiol. 2010;55:1955-61.
30.Priori SG, Napolitano C, Schwartz PJ, et al. Association of long QT syndrome loci and cardiac events among patients
treated with beta-blockers. JAMA. 2004;292:1341-4.

Page 67
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

31.Goldenberg I, Horr S, Moss AJ, et al. Risk for life-threatening cardiac events in patients with genotype-confirmed
long-QT syndrome and normal-range corrected QT intervals. J Am Coll Cardiol. 2011;57:51-9.
32.Choy AM, Lang CC, Chomsky DM, et al. Normalization of acquired QT prolongation in humans by intravenous
potassium. Circulation. 1997;96:2149-54.
33.Kannankeril P, Roden DM, Darbar D. Drug-induced long QT syndrome. Pharmacol Rev. 2010;62:760-81.
34.Zhang C, Kutyifa V, Moss AJ, et al. Long-QT syndrome and therapy for attention deficit/hyperactivity disorder. J
Cardiovasc Electrophysiol. 2015;26:1039-44.
35.Credible meds. Available at: http://www.crediblemeds.org. Accessed December 26, 2016.
36.Roden DM. Drug-induced prolongation of the QT interval. N Engl J Med. 2004;350:1013-22.
6.9.1.2. Catecholaminergic Polymorphic Ventricular Tachycardia
Recommendations for Catecholaminergic Polymorphic Ventricular Tachycardia
References that support the recommendations are summarized in Online Data Supplement 41.
COR LOE Recommendations
1. In patients with catecholaminergic polymorphic ventricular tachycardia, a beta
I B-NR
blocker is recommended (1, 2).
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

2. In patients with catecholaminergic polymorphic ventricular tachycardia and


recurrent sustained VT or syncope, while receiving adequate or maximally
I B-NR tolerated beta blocker, treatment intensification with either combination
medication therapy (e.g., beta blocker, flecainide), left cardiac sympathetic
denervation, and/or an ICD is recommended (2-6).
3. In patients with catecholaminergic polymorphic ventricular tachycardia and
IIa B-NR with clinical VT or exertional syncope, genetic counseling and genetic testing
are reasonable (7).
Figure 13

Figure 13. Exercise-Induced Polymorphic VT in Catecholaminergic Polymorphic Ventricular Tachycardia

References
1. Hayashi M, Denjoy I, Extramiana F, et al. Incidence and risk factors of arrhythmic events in catecholaminergic
polymorphic ventricular tachycardia. Circulation. 2009;119:2426-34.
2. Roston TM, Vinocur JM, Maginot KR, et al. Catecholaminergic polymorphic ventricular tachycardia in children:
analysis of therapeutic strategies and outcomes from an international multicenter registry. Circ Arrhythm
Electrophysiol. 2015;8:633-42.

Page 68
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

3. Collura CA, Johnson JN, Moir C, et al. Left cardiac sympathetic denervation for the treatment of long QT syndrome
and catecholaminergic polymorphic ventricular tachycardia using video-assisted thoracic surgery. Heart Rhythm.
2009;6:752-9.
4. Hofferberth SC, Cecchin F, Loberman D, et al. Left thoracoscopic sympathectomy for cardiac denervation in patients
with life-threatening ventricular arrhythmias. J Thorac Cardiovasc Surg. 2014;147:404-9.
5. Schneider HE, Steinmetz M, Krause U, et al. Left cardiac sympathetic denervation for the management of life-
threatening ventricular tachyarrhythmias in young patients with catecholaminergic polymorphic ventricular
tachycardia and long QT syndrome. Clin Res Cardiol. 2013;102:33-42.
6. van der Werf C, Kannankeril PJ, Sacher F, et al. Flecainide therapy reduces exercise-induced ventricular arrhythmias
in patients with catecholaminergic polymorphic ventricular tachycardia. J Am Coll Cardiol. 2011;57:2244-54.
7. Priori SG, Napolitano C, Memmi M, et al. Clinical and molecular characterization of patients with catecholaminergic
polymorphic ventricular tachycardia. Circulation. 2002;106:69-74.
6.9.1.3. Brugada Syndrome
Recommendations for Brugada Syndrome
References that support the recommendations are summarized in Online Data Supplement 42 and
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Systematic Review Report.


COR LOE Recommendations
1. In asymptomatic patients with only inducible type 1 Brugada
I B-NR electrocardiographic pattern, observation without therapy is recommended (1-
5).
2. In patients with Brugada syndrome with spontaneous type 1 Brugada
electrocardiographic pattern and cardiac arrest, sustained VA or a recent
I B-NR
history of syncope presumed due to VA, an ICD is recommended if a meaningful
survival of greater than 1 year is expected (4, 6).
3. In patients with Brugada syndrome experiencing recurrent ICD shocks for
I B-NR polymorphic VT, intensification of therapy with quinidine or catheter ablation
is recommended (7-11).
4. In patients with spontaneous type 1 Brugada electrocardiographic pattern and
I B-NR symptomatic VA who either are not candidates for or decline an ICD, quinidine
or catheter ablation is recommended (7, 9-11).
5. In patients with suspected Brugada syndrome in the absence of a spontaneous
IIa B-NR type 1 Brugada electrocardiographic pattern, a pharmacological challenge
using a sodium channel blocker can be useful for diagnosis (12-14).
6. In patients with asymptomatic Brugada syndrome and a spontaneous type 1
Brugada electrocardiographic pattern, an electrophysiological study with
IIb B-NRSR
programmed ventricular stimulation using single and double extrastimuli may
be considered for further risk stratification (1, 6, 13, 15-17).
7. In patients with suspected or established Brugada syndrome, genetic
IIb C-EO counseling and genetic testing may be useful to facilitate cascade screening of
relatives (18-20).
SR indicated systematic review.
Figures 14 and 15

Page 69
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Figure 14. Prevention of SCD in Patients With Brugada Syndrome


Documented or
suspected Brugada
syndrome

Genotyping Spontaneous Type 1 Suspected Brugada syndrome Lifestyle changes:


(Class IIb) Brugada ECG without Type I ECG 1. Avoid Brugada
aggravating drugs
2. Treat fever
Positive
Pharmacologic 3. Avoid excessive
Genetic counselling for Yes challenge alcohol
mutation specific (Class IIa) 4. Avoid cocaine
genotyping of 1 relatives
(Class I)
Cardiac arrest,
ICD
recent unexplained Yes
candidate*
syncope
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

No
No Yes

EP study for risk Quinidine or ICD


Observe
stratification catheter ablation (Class I)
without therapy
(Class IIb) (Class I)

Quinidine or
Recurrent VT,
catheter ablation
VF Storm
(Class I)

Colors correspond to Class of Recommendation in Table 1.


See Section 7.9.1.3 in the full-text guideline for discussion.
*ICD candidacy as determined by functional status,life expectancy or patient preference.
1 indicates primary; ECG, electrocardiogram; EP, electrophysiological; ICD implantable cardioverter-defibrillator; SCD,
sudden cardiac death; VT, ventricular tachycardia; and VF, ventricular fibrillation.

Figure 15. Brugada Syndrome

References
1. Casado-Arroyo R, Berne P, Rao JY, et al. Long-term trends in newly diagnosed Brugada syndrome: implications for
risk stratification. J Am Coll Cardiol. 2016;68:614-23.
2. Gehi AK, Duong TD, Metz LD, et al. Risk stratification of individuals with the Brugada electrocardiogram: a meta-
analysis. J Cardiovasc Electrophysiol. 2006;17:577-83.

Page 70
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

3. Hiraoka M, Takagi M, Yokoyama Y, et al. Prognosis and risk stratification of young adults with Brugada syndrome. J
Electrocardiol. 2013;46:279-83.
4. Priori SG, Gasparini M, Napolitano C, et al. Risk stratification in Brugada syndrome: results of the PRELUDE
(PRogrammed ELectrical stimUlation preDictive valuE) registry. J Am Coll Cardiol. 2012;59:37-45.
5. Probst V, Veltmann C, Eckardt L, et al. Long-term prognosis of patients diagnosed with Brugada syndrome: results
from the FINGER Brugada syndrome registry. Circulation. 2010;121:635-43.
6. Sroubek J, Probst V, Mazzanti A, et al. Programmed ventricular stimulation for risk stratification in the Brugada
syndrome: a pooled analysis. Circulation. 2016;133:622-30.
7. Belhassen B, Rahkovich M, Michowitz Y, et al. Management of Brugada syndrome: thirty-three-year experience
using electrophysiologically guided therapy with class 1a antiarrhythmic drugs. Circ Arrhythm Electrophysiol.
2015;8:1393-402.
8. Brugada J, Pappone C, Berruezo A, et al. Brugada syndrome phenotype elimination by epicardial substrate ablation.
Circ Arrhythm Electrophysiol. 2015;8:1373-81.
9. Nademanee K, Veerakul G, Chandanamattha P, et al. Prevention of ventricular fibrillation episodes in Brugada
syndrome by catheter ablation over the anterior right ventricular outflow tract epicardium. Circulation.
2011;123:1270-9.
10.Sunsaneewitayakul B, Yao Y, Thamaree S, et al. Endocardial mapping and catheter ablation for ventricular fibrillation
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

prevention in Brugada syndrome. J Cardiovasc Electrophysiol. 2012;23(suppl 1):S10-6.


11.Zhang P, Tung R, Zhang Z, et al. Characterization of the epicardial substrate for catheter ablation of Brugada
syndrome. Heart Rhythm. 2016;13:2151-8.
12.Antzelevitch C, Yan GX, Ackerman MJ, et al. J-Wave syndromes expert consensus conference report: emerging
concepts and gaps in knowledge. Europace. 2017;19:665-94.
13.Delise P, Allocca G, Marras E, et al. Risk stratification in individuals with the Brugada type 1 ECG pattern without
previous cardiac arrest: usefulness of a combined clinical and electrophysiologic approach. Eur Heart J. 2011;32:169-
76.
14.Somani R, Krahn AD, Healey JS, et al. Procainamide infusion in the evaluation of unexplained cardiac arrest: from the
Cardiac Arrest Survivors with Preserved Ejection Fraction Registry (CASPER). Heart Rhythm. 2014;11:1047-54.
15.Kusumoto FM, Bailey KR, Chaouki AS, et al. Systematic review for the 2017 AHA/ACC/HRS guideline for management
of patients with ventricular arrhythmias and the prevention of sudden cardiac death: a report of the American
College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm
Society [published online ahead of print October 30, 2017]. Circulation. doi: 10.1161/CIR.0000000000000550.
16.Sieira J, Ciconte G, Conte G, et al. Asymptomatic Brugada syndrome: clinical characterization and long-term
prognosis. Circ Arrhythm Electrophysiol. 2015;8:1144-50.
17.Sieira J, Conte G, Ciconte G, et al. Prognostic value of programmed electrical stimulation in Brugada syndrome: 20
years experience. Circ Arrhythm Electrophysiol. 2015;8:777-84.
18.Crotti L, Marcou CA, Tester DJ, et al. Spectrum and prevalence of mutations involving BrS1- through BrS12-
susceptibility genes in a cohort of unrelated patients referred for Brugada syndrome genetic testing: implications for
genetic testing. J Am Coll Cardiol. 2012;60:1410-8.
19.Probst V, Wilde AA, Barc J, et al. SCN5A mutations and the role of genetic background in the pathophysiology of
Brugada syndrome. Circ Cardiovasc Genet. 2009;2:552-7.
20.Risgaard B, Jabbari R, Refsgaard L, et al. High prevalence of genetic variants previously associated with Brugada
syndrome in new exome data. Clin Genet. 2013;84:489-95.

Page 71
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6.9.1.4. Early Repolarization J-wave Syndrome


Recommendations for Early Repolarization Syndrome
References that support the recommendations are summarized in Online Data Supplement 43.
COR LOE Recommendations
1. In asymptomatic patients with an early repolarization pattern on ECG,
I B-NR
observation without treatment is recommended (1, 2).
2. In patients with early repolarization pattern on ECG and cardiac arrest or
I B-NR
sustained VA, an ICD is recommended (3, 4).
III: No 3. In patients with early repolarization pattern on ECG, genetic testing is not
B-NR
Benefit recommended (5).

References
1. Rosso R, Kogan E, Belhassen B, et al. J-point elevation in survivors of primary ventricular fibrillation and matched
control subjects: incidence and clinical significance. J Am Coll Cardiol. 2008;52:1231-8.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

2. Adhikarla C, Boga M, Wood AD, et al. Natural history of the electrocardiographic pattern of early repolarization in
ambulatory patients. Am J Cardiol. 2011;108:1831-5.
3. Haissaguerre M, Sacher F, Nogami A, et al. Characteristics of recurrent ventricular fibrillation associated with
inferolateral early repolarization role of drug therapy. J Am Coll Cardiol. 2009;53:612-9.
4. Siebermair J, Sinner MF, Beckmann BM, et al. Early repolarization pattern is the strongest predictor of arrhythmia
recurrence in patients with idiopathic ventricular fibrillation: results from a single centre long-term follow-up over
20 years. Europace. 2016;18:718-25.
5. Sinner MF, Porthan K, Noseworthy PA, et al. A meta-analysis of genome-wide association studies of the
electrocardiographic early repolarization pattern. Heart Rhythm. 2012;9:1627-34.

6.9.1.5. Short QT Syndrome


Recommendations for Short QT Syndrome
References that support the recommendations are summarized in Online Data Supplement 44.
COR LOE Recommendations
1. In asymptomatic patients with a short QTc interval, observation without
I B-NR
treatment is recommended (1, 2).
2. In patients with short QT syndrome who have a cardiac arrest or sustained VA,
I B-NR an ICD is recommended if meaningful survival greater than 1 year is expected
(3-5).
3. In patients with short QT syndrome and recurrent sustained VA, treatment
IIa C-LD
with quinidine can be useful (3, 5, 6).
4. In patients with short QT syndrome and VT/VF storm, isoproterenol infusion
IIa C-LD
can be effective (7).
5. In patients with short QT syndrome, genetic testing may be considered to
IIb C-EO
facilitate screening of first-degree relatives (4).

References
1. Dhutia H, Malhotra A, Parpia S, et al. The prevalence and significance of a short QT interval in 18,825 low-risk
individuals including athletes. Br J Sports Med. 2016;50:124-9.
2. Gollob MH, Redpath CJ, Roberts JD. The short QT syndrome: proposed diagnostic criteria. J Am Coll Cardiol.
2011;57:802-12.
3. Giustetto C, Schimpf R, Mazzanti A, et al. Long-term follow-up of patients with short QT syndrome. J Am Coll
Cardiol. 2011;58:587-95.

Page 72
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

4. Mazzanti A, Kanthan A, Monteforte N, et al. Novel insight into the natural history of short QT syndrome. J Am Coll
Cardiol. 2014;63:1300-8.
5. Villafane J, Atallah J, Gollob MH, et al. Long-term follow-up of a pediatric cohort with short QT syndrome. J Am Coll
Cardiol. 2013;61:1183-91.
6. Giustetto C, Di MF, Wolpert C, et al. Short QT syndrome: clinical findings and diagnostic-therapeutic implications.
Eur Heart J. 2006;27:2440-7.
7. Bun SS, Maury P, Giustetto C, et al. Electrical storm in short-QT syndrome successfully treated with Isoproterenol. J
Cardiovasc Electrophysiol. 2012;23:1028-30.

7. VA in the Structurally Normal Heart


Recommendations for VA in the Structurally Normal Heart
References that support the recommendations are summarized in Online Data Supplement 45.
COR LOE Recommendation
1. In patients with symptomatic PVCs in an otherwise normal heart, treatment
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

I B-R with a beta blocker or nondihydropyridine calcium channel blocker is useful to


reduce recurrent arrhythmias and improve symptoms (1, 2).
2. In patients with symptomatic VA in an otherwise normal heart, treatment with
IIa an antiarrhythmic medication is reasonable to reduce recurrent symptomatic
B-R
arrhythmias and improve symptoms if beta blockers and nondihydropyridine
calcium channel blockers are ineffective or not tolerated (3, 4).

References
1. Gill JS, Blaszyk K, Ward DE, et al. Verapamil for the suppression of idiopathic ventricular tachycardia of left bundle
branch block-like morphology. Am Heart J. 1993;126:1126-33.
2. Gill JS, Ward DE, Camm AJ. Comparison of verapamil and diltiazem in the suppression of idiopathic ventricular
tachycardia. Pacing Clin Electrophysiol. 1992;15:2122-6.
3. Kontos MC, Diercks DB, Ho PM, et al. Treatment and outcomes in patients with myocardial infarction treated with
acute beta-blocker therapy: results from the American College of Cardiology's NCDR(R). Am Heart J. 2011;161:864-70.
4. Levine JH, Massumi A, Scheinman MM, et al. Intravenous amiodarone for recurrent sustained hypotensive
ventricular tachyarrhythmias. Intravenous Amiodarone Multicenter Trial Group. J Am Coll Cardiol. 1996;27:67-75.

7.1. Outflow Tract and Atrioventricular Annular VA


Recommendations for Outflow Tract VA
References that support the recommendations are summarized in Online Data Supplement 46.
COR LOE Recommendations
1. In patients with symptomatic outflow tract VA in an otherwise normal heart
I B-NR for whom antiarrhythmic medications are ineffective, not tolerated, or not the
patients preference, catheter ablation is useful (1-3).
2. In patients with symptomatic outflow tract VT in an otherwise normal heart,
I B-NR
a beta blocker or a calcium channel blocker is useful (1-3).

References
1. Tada H, Ito S, Naito S, et al. Idiopathic ventricular arrhythmia arising from the mitral annulus: a distinct subgroup of
idiopathic ventricular arrhythmias. J Am Coll Cardiol. 2005;45:877-86.
2. Yamada T, Litovsky SH, Kay GN. The left ventricular ostium: an anatomic concept relevant to idiopathic ventricular
arrhythmias. Circ Arrhythm Electrophysiol. 2008;1:396-404.

Page 73
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

3. Yamada T, Maddox WR, McElderry HT, et al. Radiofrequency catheter ablation of idiopathic ventricular arrhythmias
originating from intramural foci in the left ventricular outflow tract: efficacy of sequential versus simultaneous unipolar
catheter ablation. Circ Arrhythm Electrophysiol. 2015;8:344-52.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Page 74
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

7.2. Papillary Muscle VA


Recommendation for Papillary Muscle VA (PVCs and VT)
References that support the recommendation are summarized in Online Data Supplement 47.
COR LOE Recommendation
1. In patients with symptomatic VA arising from the papillary muscles for whom
I B-NR antiarrhythmic medications are ineffective, not tolerated, or not the patients
preference, catheter ablation is useful (1-5).

References
1. Ban JE, Lee HS, Lee DI, et al. Electrophysiological characteristics related to outcome after catheter ablation of
idiopathic ventricular arrhythmia originating from the papillary muscle in the left ventricle. Korean Circ J.
2013;43:811-8.
2. Crawford T, Mueller G, Good E, et al. Ventricular arrhythmias originating from papillary muscles in the right
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

ventricle. Heart Rhythm. 2010;7:725-30.


3. Doppalapudi H, Yamada T, McElderry HT, et al. Ventricular tachycardia originating from the posterior papillary
muscle in the left ventricle: a distinct clinical syndrome. Circ Arrhythm Electrophysiol. 2008;1:23-9.
4. Yamada T, Doppalapudi H, McElderry HT, et al. Electrocardiographic and electrophysiological characteristics in
idiopathic ventricular arrhythmias originating from the papillary muscles in the left ventricle: relevance for catheter
ablation. Circ Arrhythm Electrophysiol. 2010;3:324-31.
5. Yokokawa M, Good E, Desjardins B, et al. Predictors of successful catheter ablation of ventricular arrhythmias arising
from the papillary muscles. Heart Rhythm. 2010;7:1654-9.

7.3. Interfascicular Reentrant VT (Belhassen Tachycardia)


Recommendations for Interfascicular Reentrant VT (Belhassen Tachycardia)
References that support the recommendations are summarized in Online Data Supplement 48
COR LOE Recommendations
1. In patients with verapamil-sensitive, idiopathic LVT related to interfascicular
I B-NR reentry for whom antiarrhythmic medications are ineffective, not tolerated,
or not the patients preference, catheter ablation is useful (1-3).
2. In patients with sustained hemodynamically tolerated verapamil-sensitive,
I B-NR idiopathic LVT related to interfascicular reentry, intravenous verapamil is
recommended for VT termination (3-6).
3. In patients with recurrent verapamil-sensitive idiopathic LVT, chronic therapy
IIa C-LD
with oral verapamil can be useful (7-10).

References
1. Lin D, Hsia HH, Gerstenfeld EP, et al. Idiopathic fascicular left ventricular tachycardia: linear ablation lesion strategy
for noninducible or nonsustained tachycardia. Heart Rhythm. 2005;2:934-9.
2. Liu Y, Fang Z, Yang B, et al. Catheter ablation of fascicular ventricular tachycardia: long-term clinical outcomes and
mechanisms of recurrence. Circ Arrhythm Electrophysiol. 2015;8:1443-51.
3. Nogami A, Naito S, Tada H, et al. Demonstration of diastolic and presystolic Purkinje potentials as critical potentials
in a macroreentry circuit of verapamil-sensitive idiopathic left ventricular tachycardia. J Am Coll Cardiol.
2000;36:811-23.
4. Belhassen B, Rotmensch HH, Laniado S. Response of recurrent sustained ventricular tachycardia to verapamil. Br
Heart J. 1981;46:679-82.
5. German LD, Packer DL, Bardy GH, et al. Ventricular tachycardia induced by atrial stimulation in patients without
symptomatic cardiac disease. Am J Cardiol. 1983;52:1202-7.

Page 75
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

6. Tsuchiya T, Okumura K, Honda T, et al. Effects of verapamil and lidocaine on two components of the re-entry circuit
of verapamil-senstitive idiopathic left ventricular tachycardia. J Am Coll Cardiol. 2001;37:1415-21.
7. Anderson JH, Tester DJ, Will ML, et al. Whole-exome molecular autopsy after exertion-related sudden unexplained
death in the young. Circ Cardiovasc Genet. 2016;9:259-65.
8. Ohe T, Shimomura K, Aihara N, et al. Idiopathic sustained left ventricular tachycardia: clinical and electrophysiologic
characteristics. Circulation. 1988;77:560-8.
9. Snyder C, Bishara J, Darling R, et al. Verapamil-sensitive ventricular tachycardia in an infant. Congenit Heart Dis.
2006;1:124-6.
10.Wang JD, Fu YC, Jan SL, et al. Verapamil sensitive idiopathic ventricular tachycardia in an infant. Jpn Heart J.
2003;44:667-71.

7.4. Idiopathic Polymorphic VT/VF


Recommendations for Idiopathic Polymorphic VT/VF
References that support the recommendations are summarized in Online Data Supplement 49.
COR LOE Recommendations
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

1. In young patients (<40 years of age) with unexplained SCA, unexplained near
drowning, or recurrent exertional syncope, who do not have ischemic or other
I B-NR
structural heart disease, further evaluation for genetic arrhythmia syndromes
is recommended (1-8).
2. In patients resuscitated from SCA due to idiopathic polymorphic VT or VF, an
I B-NR ICD is recommended if meaningful survival greater than 1 year is expected (9-
13).
3. For patients with recurrent episodes of idiopathic VF initiated by PVCs with a
I B-NR
consistent QRS morphology, catheter ablation is useful (11, 14).

References
1. Anderson JH, Tester DJ, Will ML, et al. Whole-exome molecular autopsy after exertion-related sudden unexplained
death in the young. Circ Cardiovasc Genet. 2016;9:259-65.
2. Dalal A, Czosek RJ, Kovach J, et al. Clinical presentation of pediatric patients at risk for sudden cardiac arrest. J
Pediatr. 2016;177:191-6.
3. Kumar S, Peters S, Thompson T, et al. Familial cardiological and targeted genetic evaluation: low yield in sudden
unexplained death and high yield in unexplained cardiac arrest syndromes. Heart Rhythm. 2013;10:1653-60.
4. Linzer M, Pritchett EL, Pontinen M, et al. Incremental diagnostic yield of loop electrocardiographic recorders in
unexplained syncope. Am J Cardiol. 1990;66:214-9.
5. Solomon SD, Zelenkofske S, McMurray JJ, et al. Sudden death in patients with myocardial infarction and left
ventricular dysfunction, heart failure, or both. N Engl J Med. 2005;352:2581-8.
6. Tester DJ, Medeiros-Domingo A, Will ML, et al. Unexplained drownings and the cardiac channelopathies: a
molecular autopsy series. Mayo Clin Proc. 2011;86:941-7.
7. Tzimas I, Zingraf JC, Bajanowski T, et al. The role of known variants of KCNQ1, KCNH2, KCNE1, SCN5A, and NOS1AP
in water-related deaths. Int J Legal Med. 2016;130:1575-9.
8. Wang D, Shah KR, Um SY, et al. Cardiac channelopathy testing in 274 ethnically diverse sudden unexplained deaths.
Forensic Sci Int. 2014;237:90-9.
9. Conte G, Caputo ML, Regoli F, et al. True idiopathic ventricular fibrillation in out-of-hospital cardiac arrest survivors
in the Swiss Canton Ticino: prevalence, clinical features, and long-term follow-up. Europace. 2017;19:259-66.
10.Frommeyer G, Dechering DG, Kochhauser S, et al. Long-time "real-life" performance of the subcutaneous ICD in
patients with electrical heart disease or idiopathic ventricular fibrillation. J Interv Card Electrophysiol. 2016;47:185-
8.
11.Hassaguerre M, Shoda M, Jais P, et al. Mapping and ablation of idiopathic ventricular fibrillation. Circulation.
2002;106:962-7.
12.Knecht S, Sacher F, Wright M, et al. Long-term follow-up of idiopathic ventricular fibrillation ablation: a multicenter
study. J Am Coll Cardiol. 2009;54:522-8.

Page 76
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

13.Leenhardt A, Glaser E, Burguera M, et al. Short-coupled variant of torsade de pointes. A new electrocardiographic
entity in the spectrum of idiopathic ventricular tachyarrhythmias. Circulation. 1994;89:206-15.
14.Hassaguerre M, Shah DC, Jais P, et al. Role of Purkinje conducting system in triggering of idiopathic ventricular
fibrillation. Lancet. 2002;359:677-8.

8. PVC-Induced Cardiomyopathy
Recommendations for PVC-Induced Cardiomyopathy
References that support the recommendations are summarized in Online Data Supplement 50.
COR LOE Recommendations
1. For patients who require arrhythmia suppression for symptoms or declining
ventricular function suspected to be due to frequent PVCs (generally >15% of
I B-NR beats and predominately of 1 morphology) and for whom antiarrhythmic
medications are ineffective, not tolerated, or not the patients preference,
catheter ablation is useful (1, 2).
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

2. In patients with PVC-induced cardiomyopathy, pharmacologic treatment (e.g.,


IIa B-NR beta blocker, amiodarone) is reasonable to reduce recurrent arrhythmias, and
improve symptoms and LV function (3, 4).

References
1. Hassaguerre M, Shah DC, Jais P, et al. Role of Purkinje conducting system in triggering of idiopathic ventricular
fibrillation. Lancet. 2002; 359:677-8.
2. Hassaguerre M, Shoda M, Jais P, et al. Mapping and ablation of idiopathic ventricular fibrillation. Circulation.
2002;106:962-7.
3. Lee GK, Klarich KW, Grogan M, et al. Premature ventricular contraction-induced cardiomyopathy: a treatable
condition. Circ Arrhythm Electrophysiol. 2012;5:229-36.
4. Singh SN, Fletcher RD, Fisher SG, et al. Amiodarone in patients with congestive heart failure and asymptomatic
ventricular arrhythmia. Survival Trial of Antiarrhythmic Therapy in Congestive Heart Failure. N Engl J Med.
1995;333:77-82.

9. VA and SCD Related to Specific Populations


9.1. Pregnancy
Recommendations for Pregnancy
References that support the recommendations are summarized in Online Data Supplement 51.
COR LOE Recommendations
1. In mothers with long QT syndrome, a beta blocker should be continued during
I B-NR pregnancy and throughout the postpartum period including in women who are
breastfeeding (1).
2. In the pregnant patient with sustained VA, electrical cardioversion is safe and
I C-EO
effective and should be used with standard electrode configuration (2, 3).
3. In pregnant patients needing an ICD or VT ablation, it is reasonable to undergo
IIa B-NR
these procedures during pregnancy, preferably after the first trimester (4, 5).

References
1. Seth R, Moss AJ, McNitt S, et al. Long QT syndrome and pregnancy. J Am Coll Cardiol. 2007;49:1092-8.

Page 77
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

2. Jeejeebhoy FM, Zelop CM, Lipman S, et al. Cardiac arrest in pregnancy: a scientific statement from the American
Heart Association. Circulation. 2015;132:1747-73.
3. Vanden Hoek TL, Morrison LJ, Shuster M, et al. Part 12: cardiac arrest in special situations: 2010 American Heart
Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Circulation.
2010;122(suppl 3):S829-61.
4. Natale A, Davidson T, Geiger MJ, et al. Implantable cardioverter-defibrillators and pregnancy: a safe combination?
Circulation. 1997;96:2808-12.
5. Colletti PM, Lee KH, Elkayam U. Cardiovascular imaging of the pregnant patient. Am J Roentgenol. 2013;200:515-21.

9.2. Older Patients With Comorbidities


Recommendation for Older Patients With Comorbidities
See Systematic Review Report (1).
COR LOE Recommendation
1. For older patients and those with significant comorbidities, who meet
IIa B-NRSR indications for a primary prevention ICD, an ICD is reasonable if meaningful
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

survival of greater than 1 year is expected (1).


SR indicates systematic review.

Reference
1. Kusumoto FM, Bailey KR, Chaouki AS, et al. Systematic review for the 2017 AHA/ACC/HRS guideline for management
of patients with ventricular arrhythmias and the prevention of sudden cardiac death: a report of the American
College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm
Society [published online ahead of print October 30, 2017]. Circulation. doi: 10.1161/CIR.0000000000000550.

Page 78
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

9.3. Medication-Induced Arrhythmias


Recommendations for Medication-Induced Arrhythmias
References that support the recommendations are summarized in Online Data Supplement 52 and 53.
Digoxin
COR LOE Recommendation
1. Administration of digoxin antibodies is recommended for patients who present
I B-NR
with sustained VA potentially due to digoxin toxicity (1, 2).
Medication-Induced QT Prolongation and Torsades de Pointes
COR LOE Recommendations
2. In patients with recurrent torsades de pointes associated with acquired QT
prolongation and bradycardia that cannot be suppressed with intravenous
I B-NR
magnesium administration, increasing the heart rate with atrial or ventricular
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

pacing or isoproterenol are recommended to suppress the arrhythmia (3).


3. For patients with QT prolongation due to a medication, hypokalemia,
hypomagnesemia, or other acquired factor and recurrent torsades de pointes,
I C-LD
administration of intravenous magnesium sulfate is recommended to suppress
the arrhythmia (4, 5).
4. For patients with torsades de pointes associated with acquired QT
I C-LD prolongation, potassium repletion to 4.0 mmol per L or more and magnesium
repletion to normal values (e.g., 2.0 mmol/L) are beneficial (6, 7).
Sodium Channel BlockerRelated Toxicity
COR LOE Recommendations
5. In patients taking sodium channel blockers who present with elevated
defibrillation or pacing thresholds, discontinuing the presumed responsible
IIa C-LD
medication or reprogramming the device can be useful to restore effective
device therapy (8, 9).
6. In patients with congenital or acquired long QT syndrome, QT-prolonging
III: Harm B-NR
medications are potentially harmful (10).
Table 10

References
1. Antman EM, Wenger TL, Butler VP Jr, et al. Treatment of 150 cases of life-threatening digitalis intoxication with
digoxin-specific Fab antibody fragments. Final report of a multicenter study. Circulation. 1990;81:1744-52.
2. Chan BS, Buckley NA. Digoxin-specific antibody fragments in the treatment of digoxin toxicity. Clin Toxicol.
2014;52:824-36.
3. Keren A, Tzivoni D, Gavish D, et al. Etiology, warning signs and therapy of torsade de pointes. A study of 10 patients.
Circulation. 1981;64:1167-74.
4. Tzivoni D, Banai S, Schuger C, et al. Treatment of torsade de pointes with magnesium sulfate. Circulation.
1988;77:392-7.
5. Kannankeril P, Roden DMDarbar D. Drug-induced long QT syndrome. Pharmacol Rev. 2010;62:760-81.
6. Choy AM, Lang CC, Chomsky DM, et al. Normalization of acquired QT prolongation in humans by intravenous
potassium. Circulation. 1997;96:2149-54.
7. Yang T, Roden DM. Extracellular potassium modulation of drug block of IKr. Implications for torsade de pointes and
reverse use-dependence. Circulation. 1996;93:407-11.
8. Hellestrand KJ, Burnett PJ, Milne JR, et al. Effect of the antiarrhythmic agent flecainide acetate on acute and chronic
pacing thresholds. Pacing Clin Electrophysiol. 1983;6:892-9.

Page 79
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

9. Echt DS, Black JN, Barbey JT, et al. Evaluation of antiarrhythmic drugs on defibrillation energy requirements in dogs.
Sodium channel block and action potential prolongation. Circulation. 1989;79:1106-17.
10.Schwartz PJ, Woosley RL. Predicting the unpredictable: drug-induced QT prolongation and torsades de pointes. J Am
Coll Cardiol. 2016;67:1639-50.

9.4. Adult Congenital Heart Disease


Recommendations for Adult Congenital Heart Disease
References that support the recommendations are summarized in Online Data Supplement 54.
COR LOE Recommendations
1. Adult patients with repaired complex congenital heart disease presenting with
I B-NR frequent, complex, or sustained VA, or unexplained syncope should undergo
evaluation for potential residual anatomic or coronary abnormalities (1-6).
2. In patients with adult congenital heart disease and complex or sustained VA in
the presence of important residual hemodynamic lesions, treatment of
I B-NR
hemodynamic abnormalities with catheter or surgical intervention as feasible
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

is indicated prior to consideration of ablation or an ICD (3, 7-12).


3. In patients with adult congenital heart disease and hemodynamically unstable
VT, an ICD is recommended after evaluation and appropriate treatment for
I B-NR
residual lesions/ventricular dysfunction if meaningful survival of greater than
1 year is expected (13-17).
4. In patients with adult congenital heart disease with SCA due to VT or VF in the
I B-NR absence of reversible causes, an ICD is recommended if meaningful survival of
greater than 1 year is expected (13-17).
5. In adults with repaired tetralogy of Fallot physiology with high-risk
IIa B-NR characteristics and frequent VA, an electrophysiological study can be useful to
evaluate the risk of sustained VT/VF (18, 19).
6. In adults with repaired tetralogy of Fallot physiology and inducible VT/VF or
IIa B-NR
spontaneous sustained VT, implantation of an ICD is reasonable (1, 19, 20).
7. In patients with adult congenital heart disease with recurrent sustained
IIa B-NR monomorphic VT or recurrent ICD shocks for VT, catheter ablation can be
effective (21-25).
8. In adults with repaired severe complexity adult congenital heart disease and
IIa B-NR frequent or complex VA, a beta blocker can be beneficial to reduce the risk of
SCA (26).
9. In patients with repaired moderate or severe complexity adult congenital heart
disease with unexplained syncope and at least moderate ventricular
dysfunction or marked hypertrophy, either ICD implantation or an
IIa B-NR
electrophysiological study with ICD implantation for inducible sustained VA is
reasonable if meaningful survival of greater than 1 year is expected (5, 16, 27-
29).
10. In patients with adult congenital heart disease and severe ventricular
dysfunction (LVEF <35%) and symptoms of heart failure despite GDMT or
IIb B-NR
additional risk factors, ICD implantation may be considered if meaningful
survival of greater than 1 year is expected (14-16, 20).
11. In patients with adult congenital heart disease who have asymptomatic VA,
III:
B-NR prophylactic antiarrhythmic therapy with class Ic medications (i.e., flecainide,
Harm
propafenone) or amiodarone is potentially harmful (30-32).

Page 80
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Table 11 and Figure 16

Table 11. Congenital Heart Disease: Risk Factors for VA/SCD


Incidence Incidence of
Congenital Heart Disease Higher Risk Characteristics
of VA SCD
Simple complexity
ASD 2%6% <1.5% Ventricular pacing
(44, 47, 57-62) RV dilatation
VSD 3%18% <3% Pulmonary hypertension
(27, 44, 47, 57-63) NKX2.5 gene
Moderate complexity
Tetralogy of Fallot 14%31% 1.4%8.3% Unexplained syncope
(1, 2, 5, 6, 28, 34, 36, 44, 46, 47, 54-56, Frequent or complex VA
62-65) Sustained VT
QRS duration 180 ms
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Inducible sustained VT
Atrial tachycardia
Decreased LVEF
Dilated right ventricle
Severe PR
Severe PS
Aortic stenosis 10%34% 3%20% Unexplained syncope
(27, 44, 56) Severe LV hypertrophy
Aortic stenosis mean pressure gradient
>40 mm Hg
Ventricular dysfunction
Coarctation of aorta 2% 2% Aneurysm at repair site
(28, 29, 44, 46, 56, 62) Aortic stenosis
Systemic hypertension
Premature coronary artery disease
Ebsteins anomaly 2% 3%6% Cardiomegaly
(45, 47, 55) Atrial fibrillation
Wide complex tachycardia
Mitral regurgitation
Dilated RVOT
Severe complexity
Transposition of the great arteries Atrial switch
(27, 44-48, 51, 55, 56, 62) Mustard repair
Atrial switch 2% 3%9.5% Prior VSD closure
Unexplained syncope
Arterial switch 2% 1% Atrial tachycardia
Coronary orifice stenosis
cc-TGA 10% 17%25% Systemic ventricular dysfunction
Severe tricuspid regurgitation
Truncus arteriosus 10% 4% Multiple surgical repairs
(66, 67) Coronary anomalies
Ventricular dysfunction and/or
hypertrophy
Fontan repair for univentricular 5%17% 2.8%5.4% Atrial tachycardia
physiology* Longer duration of follow-up
(27, 37, 44, 45, 47, 55, 68) Ascites
Protein-losing enteropathy

Page 81
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

*Univentricular physiology includes: Tricuspid atresia, Double inlet left ventricle, Mitral atresia, Hypoplastic left heart,
Unbalanced AV septal defect.
ASD indicates atrial septal defect; cc-TGA, congenitally corrected transposition of the great arteries; LV, left ventricular;
LVEF, left ventricular ejection fraction; PR, pulmonary regurgitation; PS, pulmonary stenosis; RV, right ventricular; RVOT,
right ventricular outflow tract; SCD, sudden cardiac death; VA, ventricular arrhythmia; VSD, ventricular septal defect; and
VT, ventricular tachycardia.

Figure 16. Prevention of SCD in Patients With Adult Congenital Heart Disease
ACHD with documented or
suspected life-threatening VA

Evaluation and
treatment (if appropriate)
for residual anatomic or
coronary abnormalities
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

(Class I)

Initial presentation: ACHD with high-risk Adults with repaired TOF


resuscitated features* and frequent or complex VA
cardiac arrest and frequent VA

ICD EP study Sustained VT


(Class I) (Class IIa)
Beta blocker
Inducible (Class IIa)
sustained VT/VF Unexplained syncope
and at least moderate
Recurrent ICD ventricular dysfunction or
sustained VT (Class IIa) marked hypertrophy

Catheter
ablation
(Class IIa)
EP study ICD
(Class IIa) (Class IIa)

If positive
Colors correspond to Class of Recommendation in Table 1.
See Section 10.8 in the full-text guideline for discussion.
*High-risk features: prior palliative systemic to pulmonary shunts, unexplained syncope, frequent PVC, atrial tachycardia,
QRS duration 180 ms, decreased LVEF or diastolic dysfunction, dilated right ventricle, severe pulmonary regurgitation
or stenosis, or elevated levels of BNP.
Frequent VA refers to frequent PVCs and/or nonsustained VT.
ACHD indicates adult congenital heart disease; BNP, B-type natriuretic peptide; EP, electrophysiological; ICD, implantable
cardioverter-defibrillator; LVEF, left ventricular ejection fraction; PVC, premature ventricular complexes; SCD, sudden
cardiac death; TOF, tetralogy of Fallot; VA, ventricular arrhythmia; and VT, ventricular tachycardia.

References
1. Diller GP, Kempny A, Liodakis E, et al. Left ventricular longitudinal function predicts life-threatening ventricular
arrhythmia and death in adults with repaired tetralogy of Fallot. Circulation. 2012;125:2440-6.

Page 82
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

2. Gatzoulis MA, Till JA, Somerville J, et al. Mechanoelectrical interaction in tetralogy of Fallot. QRS prolongation
relates to right ventricular size and predicts malignant ventricular arrhythmias and sudden death. Circulation.
1995;92:231-7.
3. Harrison DA, Harris L, Siu SC, et al. Sustained ventricular tachycardia in adult patients late after repair of tetralogy of
Fallot. J Am Coll Cardiol. 1997;30:1368-73.
4. Knauth AL, Gauvreau K, Powell AJ, et al. Ventricular size and function assessed by cardiac MRI predict major adverse
clinical outcomes late after tetralogy of Fallot repair. Heart. 2008;94:211-6.
5. Koyak Z, de Groot JR, Bouma BJ, et al. Sudden cardiac death in adult congenital heart disease: can the unpredictable
be foreseen? Europace. 2017;19:401-6.
6. Koyak Z, Harris L, de Groot JR, et al. Sudden cardiac death in adult congenital heart disease. Circulation.
2012;126:1944-54.
7. Adamson L, Vohra HA, Haw MP. Does pulmonary valve replacement post repair of tetralogy of Fallot improve right
ventricular function? Interact Cardiovasc Thorac Surg. 2009;9:520-7.
8. Deal BJ, Scagliotti D, Miller SM, et al. Electrophysiologic drug testing in symptomatic ventricular arrhythmias after
repair of tetralogy of Fallot. Am J Cardiol. 1987;59:1380-5.
9. Khairy P, Van Hare GF, Balaji S, et al. PACES/HRS Expert consensus statement on the recognition and management
of arrhythmias in adult congenital heart disease: developed in partnership between the Pediatric and Congenital
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Heart Rhythm. 2014;11:e102-65.
10.Sabate Rotes A, Connolly HM, Warnes CA, et al. Ventricular arrhythmia risk stratification in patients with tetralogy of
Fallot at the time of pulmonary valve replacement. Circ Arrhythm Electrophysiol. 2015;8:110-6.
11.Therrien J, Provost Y, Merchant N, et al. Optimal timing for pulmonary valve replacement in adults after tetralogy of
Fallot repair. Am J Cardiol. 2005;95:779-82.
12.Therrien J, Siu SC, Harris L, et al. Impact of pulmonary valve replacement on arrhythmia propensity late after repair
of tetralogy of Fallot. Circulation. 2001;103:2489-94.
13.Kella DK, Merchant FM, Veledar E, et al. Lesion-specific differences for implantable cardioverter defibrillator
therapies in adults with congenital heart disease. Pacing Clin Electrophysiol. 2014;37:1492-8.
14.Koyak Z, de Groot JR, Van Gelder IC, et al. Implantable cardioverter defibrillator therapy in adults with congenital
heart disease: who is at risk of shocks? Circ Arrhythm Electrophysiol. 2012;5:101-10.
15.Santharam S, Hudsmith L, Thorne S, et al. Long-term follow-up of implantable cardioverter-defibrillators in adult
congenital heart disease patients: indications and outcomes. Europace. 2017;19:407-13.
16.Vehmeijer JT, Brouwer TF, Limpens J, et al. Implantable cardioverter-defibrillators in adults with congenital heart
disease: a systematic review and meta-analysis. Eur Heart J. 2016;37:1439-48.
17.Yap SC, Roos-Hesselink JW, Hoendermis ES, et al. Outcome of implantable cardioverter defibrillators in adults with
congenital heart disease: a multi-centre study. Eur Heart J. 2007;28:1854-61.
18.Chandar JS, Wolff GS, Garson A Jr, et al. Ventricular arrhythmias in postoperative tetralogy of Fallot. Am J Cardiol.
1990;65:655-61.
19.Khairy P, Landzberg MJ, Gatzoulis MA, et al. Value of programmed ventricular stimulation after tetralogy of Fallot
repair: a multicenter study. Circulation. 2004;109:1994-2000.
20.Khairy P, Harris L, Landzberg MJ, et al. Implantable cardioverter-defibrillators in tetralogy of Fallot. Circulation.
2008;117:363-70.
21.Kapel GF, Reichlin T, Wijnmaalen AP, et al. Re-entry using anatomically determined isthmuses: a curable ventricular
tachycardia in repaired congenital heart disease. Circ Arrhythm Electrophysiol. 2015;8:102-9.
22.Kapel GF, Reichlin T, Wijnmaalen AP, et al. Left-sided ablation of ventricular tachycardia in adults with repaired
tetralogy of Fallot: a case series. Circ Arrhythm Electrophysiol. 2014;7:889-97.
23.Kapel GF, Sacher F, Dekkers OM, et al. Arrhythmogenic anatomical isthmuses identified by electroanatomical
mapping are the substrate for ventricular tachycardia in repaired tetralogy of Fallot. Eur Heart J. 2017;38:268-76.
24.van Zyl M, Kapa S, Padmanabhan D, et al. Mechanism and outcomes of catheter ablation for ventricular tachycardia
in adults with repaired congenital heart disease. Heart Rhythm. 2016;13:1449-54.
25.Zeppenfeld K, Schalij MJ, Bartelings MM, et al. Catheter ablation of ventricular tachycardia after repair of congenital
heart disease: electroanatomic identification of the critical right ventricular isthmus. Circulation. 2007;116:2241-52.
26.Khairy P, Harris L, Landzberg MJ, et al. Sudden death and defibrillators in transposition of the great arteries with
intra-atrial baffles: a multicenter study. Circ Arrhythm Electrophysiol. 2008;1:250-7.
27.Engelings CC, Helm PC, Abdul-Khaliq H, et al. Cause of death in adults with congenital heart disease - an analysis of
the German National Register for Congenital Heart Defects. Int J Cardiol. 2016;211:31-6.

Page 83
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

28.Gallego P, Gonzalez AE, Sanchez-Recalde A, et al. Incidence and predictors of sudden cardiac arrest in adults with
congenital heart defects repaired before adult life. Am J Cardiol. 2012;110:109-17.
29.Tutarel O, Kempny A, Alonso-Gonzalez R, et al. Congenital heart disease beyond the age of 60: emergence of a new
population with high resource utilization, high morbidity, and high mortality. Eur Heart J. 2014;35:725-32.
30.Fish FA, Gillette PC, Benson DW Jr. Proarrhythmia, cardiac arrest and death in young patients receiving encainide
and flecainide. The Pediatric Electrophysiology Group. J Am Coll Cardiol. 1991;18:356-65.
31.Stan MN, Sathananthan M, Warnes C, et al. Amiodarone-induced thyrotoxicosis in adults with congenital heart
disease-clinical presentation and response to therapy. Endocr Pract. 2014;21:33-40.
32.Thorne SA, Barnes I, Cullinan P, et al. Amiodarone-associated thyroid dysfunction: risk factors in adults with
congenital heart disease. Circulation. 1999;100:149-54.

10. Defibrillators Other than Transvenous ICDs


10.1. Subcutaneous Implantable Cardioverter-Defibrillator
Recommendations for Subcutaneous Implantable Cardioverter-Defibrillator
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

References that support the recommendations are summarized in Online Data Supplement 55.
COR LOE Recommendations
1. n patients who meet criteria for an ICD who have inadequate vascular access
or are at high risk for infection, and in whom pacing for bradycardia or VT
I B-NR
termination or as part of CRT is neither needed nor anticipated, a
subcutaneous implantable cardioverter-defibrillator is recommended (1-5).
2. In patients who meet indication for an ICD, implantation of a subcutaneous
IIa B-NR implantable cardioverter-defibrillator is reasonable if pacing for bradycardia
or VT termination or as part of CRT is neither needed nor anticipated (1-4).
3. In patients with an indication for bradycardia pacing or CRT, or for whom
III: Harm B-NR antitachycardia pacing for VT termination is required, a subcutaneous
implantable cardioverter-defibrillator should not be implanted (1-4, 6-8).

References
1. Bardy GH, Smith WM, Hood MA, et al. An entirely subcutaneous implantable cardioverter-defibrillator. N Engl J
Med. 2010;363:36-44.
2. Weiss R, Knight BP, Gold MR, et al. Safety and efficacy of a totally subcutaneous implantable-cardioverter
defibrillator. Circulation. 2013;128:944-53.
3. Lambiase PD, Barr C, Theuns DA, et al. Worldwide experience with a totally subcutaneous implantable defibrillator:
early results from the EFFORTLESS S-ICD Registry. Eur Heart J. 2014;35:1657-65.
4. Burke MC, Gold MR, Knight BP, et al. Safety and efficacy of the totally subcutaneous implantable defibrillator: 2-year
results from a pooled analysis of the IDE study and EFFORTLESS registry. J Am Coll Cardiol. 2015;65:1605-15.
5. El-Chami MF, Levy M, Kelli HM, et al. Outcome of subcutaneous implantable cardioverter defibrillator implantation
in patients with end-stage renal disease on dialysis. J Cardiovasc Electrophysiol. 2015;26:900-4.
6. de Bie MK, Thijssen J, van Rees JB, et al. Suitability for subcutaneous defibrillator implantation: results based on data
from routine clinical practice. Heart. 2013;99:1018-23.
7. Olde Nordkamp LR, Dabiri AL, Boersma LV, et al. The entirely subcutaneous implantable cardioverter-defibrillator:
initial clinical experience in a large Dutch cohort. J Am Coll Cardiol. 2012;60:1933-9.
8. Kbe J, Reinke F, Meyer C, et al. Implantation and follow-up of totally subcutaneous versus conventional
implantable cardioverter-defibrillators: a multicenter case-control study. Heart Rhythm. 2013;10:29-36.

Page 84
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

10.2. Wearable Cardioverter-Defibrillator


Recommendations for Wearable Cardioverter-Defibrillator
References that support the recommendations are summarized in Online Data Supplement 56.
COR LOE Recommendations
1. In patients with an ICD and a history of SCA or sustained VA in whom removal
IIa B-NR of the ICD is required (as with infection), the wearable cardioverter-
defibrilator is reasonable for the prevention of SCD (1-4).
2. In patients at an increased risk of SCD but who are not ineligible for an ICD,
such as awaiting cardiac transplant, having an LVEF of 35% or less and are
within 40 days from an MI, or have newly diagnosed NICM, revascularization
IIb B-NR
within the past 90 days, myocarditis or secondary cardiomyopathy or a
systemic infection, wearable cardioverter-defibrillator may be reasonable (1-
5).
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

References
1. Chung MK. The role of the wearable cardioverter defibrillator in clinical practice. Cardiol Clin. 2014;32:253-70.
2. Chung MK, Szymkiewicz SJ, Shao M, et al. Aggregate national experience with the wearable cardioverter-
defibrillator: event rates, compliance, and survival. J Am Coll Cardiol. 2010;56:194-203.
3. Klein HU, Meltendorf U, Reek S, et al. Bridging a temporary high risk of sudden arrhythmic death. Experience with
the wearable cardioverter defibrillator (WCD). Pacing Clin Electrophysiol. 2010;33:353-67.
4. Piccini JP Sr, Allen LA, Kudenchuk PJ, et al. Wearable cardioverter-defibrillator therapy for the prevention of sudden
cardiac death: a science advisory from the American Heart Association. Circulation. 2016;133:1715-27.
5. Ellenbogen KA, Koneru JN, Sharma PS, et al. Benefit of the wearable cardioverter-defibrillator in protecting patients
after implantable-cardioverter defibrillator explant. Results from the National Registry. JACC Clin Electrophysiol.
2017;3:243-50.

11. Special Considerations for Catheter Ablation


Recommendations for Catheter Ablation
References that support the recommendations are summarized in Online Data Supplement 57.
COR LOE Recommendations
1. In patients with bundle-branch reentrant VT, catheter ablation is useful for
I C-LD
reducing the risk of recurrent VT and ICD shocks (1-3).
2. In patients with structural heart disease who have failed endocardial catheter
IIa B-NR ablation, epicardial catheter ablation can be useful for reducing the risk of
recurrent monomorphic VT (4-6).

References
1. Blanck Z, Dhala A, Deshpande S, et al. Bundle branch reentrant ventricular tachycardia: cumulative experience in 48
patients. J Cardiovasc Electrophysiol. 1993;4:253-62.
2. Lopera G, Stevenson WG, Soejima K, et al. Identification and ablation of three types of ventricular tachycardia
involving the his-purkinje system in patients with heart disease. J Cardiovasc Electrophysiol. 2004;15:52-8.
3. Mehdirad AA, Keim S, Rist K, et al. Long-term clinical outcome of right bundle branch radiofrequency catheter
ablation for treatment of bundle branch reentrant ventricular tachycardia. Pacing Clin Electrophysiol. 1995;18:2135-
43.

Page 85
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

4. Dinov B, Fiedler L, Schonbauer R, et al. Outcomes in catheter ablation of ventricular tachycardia in dilated
nonischemic cardiomyopathy compared with ischemic cardiomyopathy: results from the Prospective Heart Centre
of Leipzig VT (HELP-VT) Study. Circulation. 2014;129:728-36.
5. Tanner H, Hindricks G, Volkmer M, et al. Catheter ablation of recurrent scar-related ventricular tachycardia using
electroanatomical mapping and irrigated ablation technology: results of the prospective multicenter Euro-VT-study.
J Cardiovasc Electrophysiol. 2010;21:47-53.
6. Marchlinski FE, Haffajee CI, Beshai JF, et al. Long-term success of irrigated radiofrequency catheter ablation of
sustained ventricular tachycardia: post-approval THERMOCOOL VT trial. J Am Coll Cardiol. 2016;67:674-83.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Page 86
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

12. Postmortem Evaluation of SCD


Recommendations for Postmortem Evaluation of SCD
References that support the recommendations are summarized in Online Data Supplement 58.
COR LOE Recommendations
1. In victims of SCD without obvious causes, a standardized cardiac-specific
I B-NR
autopsy is recommended (1, 2).
2. In first-degree relatives of SCD victims who were 40 years of age or younger,
I B-NR cardiac evaluation is recommended, with genetic counseling and genetic
testing performed as indicated by clinical findings (3).
3. In victims of SCD with an autopsy that implicates a potentially heritable
IIa B-NR cardiomyopathy or absence of structural disease, suggesting a potential
cardiac channelopathy, postmortem genetic testing is reasonable (4-7).
4. In victims of SCD with a previously identified phenotype for a genetic
arrhythmia-associated disorder, but without genotyping prior to death,
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

IIa C-LD
postmortem genetic testing can be useful for the purpose of family risk
profiling (8).

References
1. Basso C, Burke M, Fornes P, et al. Guidelines for autopsy investigation of sudden cardiac death. Virchows Arch.
2008;452:11-8.
2. de Noronha SV, Behr ER, Papadakis M, et al. The importance of specialist cardiac histopathological examination in
the investigation of young sudden cardiac deaths. Europace. 2014;16:899-907.
3. Wilde AA, Behr ER. Genetic testing for inherited cardiac disease. Nat Rev Cardiol. 2013;10:571-83.
4. Tester DJ, Medeiros-Domingo A, Will ML, et al. Cardiac channel molecular autopsy: insights from 173 consecutive
cases of autopsy-negative sudden unexplained death referred for postmortem genetic testing. Mayo Clin Proc.
2012;87:524-39.
5. Tang Y, Stahl-Herz J, Sampson BA. Molecular diagnostics of cardiovascular diseases in sudden unexplained death.
Cardiovasc Pathol. 2014;23:1-4.
6. Bagnall RD, Das KJ, Duflou J, et al. Exome analysis-based molecular autopsy in cases of sudden unexplained death in
the young. Heart Rhythm. 2014;11:655-62.
7. Bagnall RD, Weintraub RG, Ingles J, et al. A prospective study of sudden cardiac death among children and young
adults. N Engl J Med. 2016;374:2441-52.
8. Semsarian C, Ingles J. Molecular autopsy in victims of inherited arrhythmias. J Arrhythmia. 2016;32:359-65.

13. Terminal Care


Recommendations for Terminal Care
References that support the recommendations are summarized in Online Data Supplement 59.
COR LOE Recommendations
1. At the time of ICD implantation or replacement, and during advance care
I C-EO planning, patients should be informed that their ICD shock therapy can be
deactivated at any time if it is consistent with their goals and preferences.
2. In patients with refractory HF symptoms, refractory sustained VA, or nearing
I C-EO the end of life from other illness, clinicians should discuss ICD shock
deactivation and consider the patients goals and preferences.

Page 87
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

14. Shared Decision-Making


Recommendations for Shared Decision-Making
References that support the recommendations are summarized in Online Data Supplement 60.
COR LOE Recommendations
1. In patients with VA or at increased risk for SCD, clinicians should adopt a
shared decision-making approach in which treatment decisions are based not
I B-NR
only on the best available evidence, but also on the patients health goals,
preferences, and values (1-5).
2. Patients considering implantation of a new ICD or replacement of an existing
ICD for a low battery should be informed of their individual risk of SCD and
I B-NR nonsudden death from HF or noncardiac conditions and the effectiveness,
safety, and potential complications of the ICD in light of their health goals,
preferences and values (1-5).
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

References
1. Lewis KB, Stacey D, Matlock DD. Making decisions about implantable cardioverter-defibrillators from implantation
to end of life: an integrative review of patients' perspectives. Patient. 2014;7:243-60.
2. Stewart GC, Weintraub JR, Pratibhu PP, et al. Patient expectations from implantable defibrillators to prevent death
in heart failure. J Cardiac Fail. 2010;16:106-13.
3. Hauptman PJ, Chibnall JT, Guild C, et al. Patient perceptions, physician communication, and the implantable
cardioverter-defibrillator. JAMA Intern Med. 2013;173:571-7.
4. Ottenberg AL, Mueller PS, Topazian RJ, et al. "It's not broke, so let's not try to fix it": why patients decline a
cardiovascular implantable electronic device. Pacing Clin Electrophysiol. 2014;37:1306-14.
5. Yuhas J, Mattocks K, Gravelin L, et al. Patients' attitudes and perceptions of implantable cardioverter-defibrillators:
potential barriers to appropriate primary prophylaxis. Pacing Clin Electrophysiol. 2012;35:1179-87.

15. Cost and Value Considerations


The key principles of value assessment as part of clinical practice guidelines have been discussed in detail
(1). Economic outcomes of clinical management strategies can be documented empirically using the same
research designs as used in establishing clinical outcomes, including RCTs and observational comparisons. In
addition, simulation models are often used to assess the value of management strategies, because the
standard for cost-effectiveness studies is to compare life-time outcomes, and clinical studies usually have
follow-up of a few years at most. Standards for economic modeling in health care have been published by an
expert group (2).
Economic assessments of alternative management strategies for VA and prevention of SCD have
primarily evaluated ICDs, including several RCTs (3-7) and observational studies (8, 9), and simulation models
(10-14). In all studies, patients who received ICDs had higher long-term costs. The high initial cost of the ICD
device and the implantation procedure leads to higher long-term costs, because there are few, if any,
subsequent cost-savings from implanting an ICD. ICDs without resynchronization capability do not reduce
hospital readmissions, and may increase late costs due to device monitoring, complications, and
replacement. However, the cost of the device and the procedure may change significantly over time.
The trial based assessments of the cost-effectiveness of the ICD are based on 3 to 6 years of
follow-up, which is considerably shorter than the lifetime perspective that is standard in cost-effectiveness
models. Because most of the incremental cost of the ICD is incurred immediately, while most of the
potential effectiveness (life-years of survival added by the ICD) is accrued over many years, estimates of ICD
cost effectiveness based on limited trial follow-up have a systematic bias toward showing lower value. Trial

Page 88
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

based economic studies that projected long-term ICD outcomes have consistently found more favorable
cost-effectiveness ratios than estimates restricted to the duration of trial follow-up (4-7). A lifetime
simulation model applied to each major trial of primary prevention ICDs also reported consistently more
favorable estimates of cost effectiveness than the estimates based on limited trial follow-up (11). Because
the framework proposed for assessing value in ACC/AHA clinical practice guidelines uses benchmarks based
on lifetime estimates (1), we have generally relied on the model-based estimates of ICD cost-effectiveness in
applying value ratings to recommendations in this guideline.
The initial cost of an ICD device is similar regardless of the clinical indication, so variations in ICD cost
effectiveness are driven primarily by potential differences in clinical effectiveness in extending survival in
different patient populations. The effect of the years of life added by an ICD on its incremental cost-
effectiveness ratio is illustrated in Figure 17: the cost-effectiveness ratio becomes rapidly unfavorable as the
extension in survival time falls below 1 year, particularly below 0.5 year. This inverse relation strongly
suggests that the value provided by an ICD will be highest when the risk of arrhythmic death due to VT/VF is
relatively high and the risk of nonarrhythmic death (either cardiac or noncardiac) is relatively low, such that
a meaningful increase in survival can be expected from the ICD. Thus, appropriate patient selection is
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

fundamental to high value care in using the ICD to prevent SCD. It should also be recognized that, cost-
effectiveness is also influenced by the costs for the ICD and implantation procedure, which are likely to
change significantly over time.
The empirical evidence suggests that ICDs are not effective for primary prevention of SCD when
implanted early after coronary artery bypass graft (15) or an acute MI (16, 17). An analysis of individual
patient level data from 3 secondary prevention trials (18) showed a significant variation (p=0.011) in the
clinical effectiveness of ICDs between patients with an LVEF 35% (hazard ratio: 0.66) and an LVEF >35%
(hazard ratio: 1.2). Some studies and simulation models suggest that ICDs might prolong life expectancy to a
greater extent when used in higher-risk patients than in lower-risk patients (19). In contrast, there is little
evidence of variation in the effectiveness or cost-effectiveness of the ICD based on factors such as age or sex
(20). Most studies of ICD effectiveness and value have been performed on patients with reduced LV function
due to prior MI or NICM. There are few data on the effectiveness or value of an ICD for other potential
clinical indications, such as cardiac channelopathies or HCM, although studies have suggested that their
potential cost effectiveness in such patients will depend on their underlying risk of SCD, with little evidence
of value in low-risk patients (14).

Figure 17. Incremental Cost-Effectiveness of ICD by Years of Life Added* (Example)

Page 89
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

*Figure based on formula: Incremental cost-effectiveness ratio = $50,000/QALYs.


CE indicated cost effectiveness; ICD, implantable cardioverter-defibrillator; LYA, life year added; and QALYs, quality-
adjusted life-years.

References
1. Anderson JL, Heidenreich PA, Barnett PG, et al. ACC/AHA statement on cost/value methodology in clinical practice
guidelines and performance measures: a report of the American College of Cardiology/American Heart Association
Task Force on Performance Measures and Task Force on Practice Guidelines. Circulation. 2014;129:2329-45.
2. Sanders GD, Neumann PJ, Basu A, et al. Recommendations for conduct, methodological practices, and reporting of
cost-effectiveness analyses: second panel on cost-effectiveness in health and medicine. JAMA. 2016;316:1093-103.
3. Mushlin AI, Hall J, Zwanziger J, et al. The cost-effectiveness of automatic implantable cardiac defibrillators. Results
from MADIT. Circulation. 1998;97:2129-35.
4. O'Brien BJ, Connolly SJ, Goeree R, et al. Cost-effectiveness of the implantable cardioverter-defibrillator. Results from
the Canadian Implantable Defibrillator Study (CIDS). Circulation. 2001;103:1416-21.
5. Larson G, Hallstrom A, McAnulty J, et al. Cost-effectiveness of the implantable cardioverter-defibrillator versus
antiarrhythmic drugs in survivors of serious ventricular tachyarrhythmias. Results of the Antiarrhythmics Versus
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Implantable Defibrillators (AVID) Economic Analysis Substudy. Circulation. 2002;105:2049-57.


6. Zwanziger J, Hall WJ, Dick AW, et al. The cost-effectiveness of implantable cardioverter-defibrillators. Results from
the Multicenter Automatic Defibrillator Implantation Trial (MADIT)-II. J Am Coll Cardiol. 2006;47:2310-8.
7. Mark DB, Nelson CL, Anstrom KJ, et al. Cost-effectiveness of defibrillator therapy or amiodarone in chronic stable
heart failure. Results from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT). Circulation. 2006;114:135-42.
8. Weiss JP, Saynina O, McDonald KM, et al. Effectiveness and cost-effectiveness of implantable cardioverter
defibrillators in the treatment of ventricular arrhythmias among Medicare beneficiaries. Am J Med. 2002;112:519-
27.
9. Al-Khatib SM, Anstrom KJ, Eisenstein EL, et al. Clinical and economic implications of the Multicenter Automatic
Defibrillator Implantation Trial-II. Ann Intern Med. 2005;142:593-600.
10.Buxton M, Caine N, Chase D, et al. A review of the evidence on the effects and costs of implantable cardioverter
defibrillator therapy in different patient groups, and modelling of cost-effectiveness and cost-utility for these groups
in a UK context. Health Technol Assess. 2006;10:iii-iv, ix-xi, 1-164.
11.Sanders GD, Hlatky MA, Owens DK. Cost effectiveness of implantable cardioverter-defibrillators. N Engl J Med.
2005;353:1471-80.
12.Smith T, Jordaens L, Theuns DAMJ, et al. The cost-effectiveness of primary prophylactic implantable defibrillator
therapy in patients with ischaemic or non-ischaemic heart disease: a European analysis. Eur Heart J. 2013;34:211-9.
13.Cowie MR, Marshall D, Drummond M, et al. Lifetime cost-effectiveness of prophylactic implantation of cardioverter
defibrillator in patients with reduced left ventricular systolic function: results of Markov modelling in a European
population. Europace. 2008;11:716-26.
14.Goldenberg I, Moss AJ, Maron BJ, et al. Cost-effectiveness of implanted defibrillators in young people with inherited
cardiac arrhythmias. Ann Noninvasive Electrocardiol. 2005;10(suppl):67-83.
15.Bigger JT. Prophylactic use of implanted cardiac defibrillators in patients at high risk for ventricular arrhythmias after
coronary-artery bypass graft surgery. N Engl J Med. 1997;337:1569-75.
16.Hohnloser SH, Kuck KH, Dorian P, et al. Prophylactic use of an implantable cardioverter-defibrillator after acute
myocardial infarction. N Engl J Med. 2004;351:2481-8.
17.Steinbeck G, Andresen D, Seidl K, et al. Defibrillator implantation early after myocardial infarction. N Engl J Med.
2009 361:1427-36.
18.Connolly SJ, Hallstrom AP, Cappato R, et al. Meta-analysis of the implantable cardioverter defibrillator secondary
prevention trials. Eur Heart J. 2000;21:2071-8.
19.Owens DK, Sanders GD, Heidenreich PA, et al. Effect of risk stratification on cost-effectiveness of the implantable
cardioverter defibrillator. Am Heart J. 2002;144:440-8.
20.Ezekowitz JA, Rowe BH, Dryden DM, et al. Systematic review: implantable cardioverter defibrillators for adults with
left ventricular systolic dysfunction. Ann Intern Med. 2007;147:251-62.

Page 90
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

16. Quality of Life


ICD implantation has not had a significant effect on QoL in the overall population of patients enrolled in RCTs
(1-3). Several studies have, however, demonstrated that the subset of patients who receive inappropriate
ICD shocks have worse QoL than patients who have an ICD but have not had inappropriate shocks (2).
Because an ICD is designed to prevent SCD rather than to reduce symptoms, it would not be expected to
improve QoL or functional status directly, but may have indirect, negative effects in some patients due to
device complications, or indirect, positive effects in some patients due to reassurance of having a protective
device in place.

References
1. Mark DB, Anstrom KJ, Sun JL, et al. Quality of life with defibrillator therapy or amiodarone in heart failure. N Engl J
Med. 2008;359:999-1008.
2. Noyes K, Corona E, Veazie P, et al. Examination of the effect of implantable cardioverter-defibrillators on health-
related quality of life: based on results from the Multicenter Automatic Defibrillator Trial-II. Am J Cardiovasc Drugs.
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

2009;9:393-400.
3. Passman R, Subacius H, Ruo B, et al. Implantable cardioverter defibrillators and quality of life: results from the
defibrillators in nonischemic cardiomyopathy treatment evaluation study. Arch Intern Med. 2007;167:2226-32.

17. Evidence Gaps and Future Research Needs


Despite the numerous advances in risk stratification for SCD and prevention and treatment of SCD and VA,
many gaps in knowledge remain. These gaps include:
Identification of patients who are most likely to benefit from an ICD among all ICD-eligible patients. The
role of novel markers (including genetic and imaging markers) and combinations of markers should be
studied.
Characterizing the role of the ICD in patient subgroups not well-represented in the pivotal ICD trials.
Such subgroups include patients 80 years of age and those with kidney disease, especially patients with
end-stage renal disease on dialysis, or multiple comorbidities.
Methods to identify and treat patients at high individual risk for SCD who are not identified by current
ICD eligibility criteria, including those who are within 40 days of an MI.
Defining the role of the ICD in patients with HCM, ARVC, cardiac sarcoidosis, and inherited cardiac
channelopathies in prospective studies (preferably RCT).
Determining the best approach to patients due for elective ICD generator replacement due to battery
depletion, but who may now be at low risk for SCA, such as if significant LVEF improvement has
occurred.
Obtaining more data on the efficacy and effectiveness of the S-ICD, compared with transvenous ICDs
and on the extent of testing required, and its use with other novel technologies, including leadless
pacemakers.
Conducting RCTs on catheter ablation of VT in IHD, and cardiomyopathies that evaluates procedural end
points, mortality, arrhythmia suppression, QoL, and costs.
Improving identification of individuals without significant ventricular dysfunction who are at risk of SCD.
Identifying mechanisms and risk factors for SCD in patients with HFpEF.
Improving emergency response to out-of-hospital cardiac arrest.
Developing better methods for identifying and ablating the arrhythmia substrate in structural heart
disease.
Developing better risk stratification of diseases and syndromes associated with sudden death, including
IHD, NICM, ACHD, and Brugada syndrome.

Page 91
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Identifying what causes different types of LQTS, CPVT, Brugada syndrome, HCM, and ARVC and
advancing the genotype-phenotype relationships, genotype-dependent risk, and genotype-based
tailoring of therapies for patients with inherited cardiomyopathies and inherited channelopathies.
Defining the most appropriate and beneficial use of WCDs.
Developing methods to identify and treat patients at high personal risk for SCD who are not identified by
current ICD eligibility criteria
Defining the role of CMR in enhancing risk stratification for SCD
Increasing research funding in this area, through existing and new mechanisms is critically
important. Some have proposed research funding strategies that would offer business incentives to the
insurance industries, while providing support for unresolved research goals. Such approaches should be
tested.

Presidents and Staff


Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

American College of Cardiology


Mary Norine Walsh, MD, FACC, President
Shalom Jacobovitz, Chief Executive Officer
William J. Oetgen, MD, MBA, FACC, Executive Vice President, Science, Education, Quality, and Publishing
Amelia Scholtz, PhD, Publications Manager, Science, Education, Quality, and Publishing

American College of Cardiology/American Heart Association


Katherine A. Sheehan, PhD, Director, Guideline Strategy and Operations
Abdul R. Abdullah, MD, Science and Medicine Advisor
Sam Shahid, MBBS, MPH, Associate Science and Medicine Advisor

American Heart Association


John J. Warner, MD, President
Nancy Brown, Chief Executive Officer
Rose Marie Robertson, MD, FAHA, Chief Science and Medicine Officer
Gayle R. Whitman, PhD, RN, FAHA, FAAN, Senior Vice President, Office of Science Operations
Prashant Nedungadi, PhD, Associate Science and Medicine Advisor, Office of Science Operations
Jody Hundley, Production Manager, Scientific Publications, Office of Science Operations

Key Words: ACC/AHA Clinical Practice Guidelines acute coronary syndrome ambulatory ECG monitoring
antiarrhythmic drug therapy arrhythmogenic cardiomyopathy athletes cardiac electrophysiology
cardiac resynchronization therapy cardiomyopathy catheter ablation congenital heart disease CT
imaging ECG echocardiography electrophysiological testing genetic arrhythmias Guidelines
heart failure imaging implantable cardioverter-defibrillator implantable and external cardioverter
devices medication-induced arrhythmias MR imaging myocardial infarction premature ventricular
beats resuscitation sarcoidosis specific pathology (e.g., congenital heart disease, myocarditis, renal
failure) stable coronary artery disease sudden cardiac arrest sudden cardiac death torsades de
pointes ventricular fibrillation ventricular tachycardia.

Page 92
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Appendix 1. Author Relationships With Industry and Other Entities (Relevant)2017 AHA/ACC/HRS Guideline for
Management of Patients With Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death (October 2017)
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Committee Employment Consultant Speakers Ownership/ Personal Research Institutional, Expert Voting
Member Bureau Partnership/ Organizational, or Witness Recusals by
Principal Other Financial Section*
Benefit
Sana M. Duke Clinical Research None None None None None None None
Al-Khatib Institute; Duke University
(Chair) Professor of Medicine
William G. Vanderbilt University Medical St. Jude Medical Boston Biosense None None None 4.1, 4.2.2,
Stevenson CenterProfessor of Scientific Webster 4.2.3, 5, 10.1,
(Vice Chair) MedicineBrigham and 5.4, 5.6, 6, 7,
Womens HospitalDirector 8, 9 (except
of Clinical Cardiac EP 9.7), 13, 15
Michael J. Mayo ClinicProfessor of Audentes None None None Transgenomic None 4.1, 4.2.2,
Ackerman Medicine, Pediatrics, and Therapeutics (Familion) 4.2.3, 4.2.6, 5
Pharmacology; Long QT Boston Scientific Blue Ox Health (except
Syndrome/Genetic Heart Gilead Sciences Corporation 5.1.5.2, 5.5),
Rhythm Clinic and the Mayo Invitae AliveCor 6, 7, 8, 9, 10
Clinic Windland Smith Rice Medtronic StemoniX (except 10.2)
Sudden Death Genomics MyoKardia 11, 13, 15
LaboratoryDirector St. Jude Medical
William J. Dominick Feld Hyde None None None None None None None
Bryant Attorney at Law

David J. University of Pennsylvania Biosense Webster None None Biosense Acutus None 4.1, 4.2.2,
Callans Health SystemProfessor of Biotronik Webster (PI) 4.2.3, 5.3,
Medicine; Associate Director Boston Scientific Endosense (PI) 5.4, 5.5.1,
of EP Medtronic 5.6, 6, 7, 8, 9
St. Jude Medical (expect 9.7),
10 (except
10.3), 13, 15
Anne B. University at Buffalo SUNY Medtronic None None None None None 4.1, 4.2.2,
Curtis Distinguished Professor; St. Jude Medical 4.2.3, 5.1.1,
Charles and Mary Bauer 5.1.2, 5.1.3,
Professor and Chair 5.1.4, 5.2,
5.4, 5.6, 6, 7,
8, 9, 10, 12,
13, 15

Page 93
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Barbara J. Getz Professor of Cardiology None None None None None None None
Deal Feinberg School of Medicine
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Northwestern University
Timm University of Maryland Biosense None None Biosense Impulse None 4.1, 4.2
Dickfeld Professor of Medicine St. Jude Medical General Dynamics (except
Siemens Electric Siemens 4.2.6), 4.3,
5.3, 5.4, 5.6,
6, 7, 8, 9
(except 9.7),
10.1, 11, 13,
15
Anne M. University of Calgary None None None Medtronic None None 4.2, 5.2.2,
Gillis Professor of Medicine 5.3.2, 6.4.1,
6.4.2, 6.4.4,
6.5, 6.7, 7, 8,
9, 10, 11
(except 11.7),
13, 15
Christopher Duke Clinical Research AstraZeneca None None AstraZeneca GE Healthcare None 4, 5.1 (except
B. Granger Institute; Duke University Gilead Sciences GlaxoSmithKline Medtronic 5.1.5), 5.2,
Professor of Medicine; GlaxoSmithKline Janssen ZOLL Medical 5.3, 5.4, 5.6,
Director, Cardiac Care Unit Janssen Pharmaceuticals Spacelabs 6, 7, 8, 9, 12,
Pharmaceuticals Phillips 13, 15
Medtronic Medtronic
Pfizer Pfizer
Sanofi-aventis Sanofi-aventis
Mark A. Stanford University School of None None None None None None None
Hlatky MedicineProfessor of
Health and Research Policy,
and of Cardiovascular
Medicine
Stephen C. Mayo ClinicProfessor None None None None None None None
Hammill Emeritus of Medicine
Jos A. UT Southwestern Medical None None None None None None None
Joglar CenterProfessor of Internal
Medicine; Clinical Cardiac
EPFellowship Program
Director
G. Neal Kay University of Alabama at None None None None None None None
BirminghamProfessor
Emeritus

Page 94
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Michael E. University of Wisconsin None None None None None None None
Field School of Medicine and Public
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

HealthDirector, Clinical EP
and Cardiac Arrhythmia
Service, Associate Professor
of Medicine
Gregg C. Ahmanson-UCLA Amgen None None Medtronic None None 4.1, 4.2.2,
Fonarow Cardiomyopathy Center Janssen IMPROVE-HF 4.2.3, 5.1
Director; UCLA Division of Pharmaceuticals (Steering (except
CardiologyCo-Chief Medtronic Committee) 5.1.5.1), 5.2,
ZS Pharma Medtronic 5.3, 5.4, 5.6,
6, 7, 8, 9, 10,
12, 13, 15
Daniel D. University of Colorado School None None None None None None None
Matlock of MedicineAssociate
Professor of Medicine
Robert J. University of Miami Miller None None None None None None None
Myerburg School of Medicine
Professor of Medicine and
Physiology
Richard L. University of Wisconsin None None None None None None None
Page Hospital and ClinicsChair,
Department of Medicine
This table represents the relationships of committee members with industry and other entities that were determined to be relevant to this document. These relationships were reviewed and
updated in conjunction with all meetings and/or conference calls of the writing committee during the document development process. The table does not necessarily reflect relationships
with industry at the time of publication. A person is deemed to have a significant interest in a business if the interest represents ownership of 5% of the voting stock or share of the business
entity, or ownership of $5,000 of the fair market value of the business entity;or if funds received by the person from the business entity exceed 5% of the persons gross income for the
previous year. Relationships that exist with no financial benefit are also included for the purpose of transparency. Relationships in this table are modest unless otherwise noted.
According to the ACC/AHA, a person has a relevant relationship IF: a) the relationship or interest relates to the same or similar subject matter, intellectual property or asset, topic, or
issue addressed in the document;or b) the company/entity (with whom the relationship exists) makes a drug, drug class, or device addressed in the document, or makes a competing drug or
device addressed in the document;or c) the person or a member of the persons household, has a reasonable potential for financial, professional or other personal gain or loss as a result of the
issues/content addressed in the document.

*Writing committee members are required to recuse themselves from voting on sections to which their specific relationships with industry and other entities may apply. Section numbers
pertain to those in the full-text guideline.
Significant relationship.
No financial benefit.

ACC indicates American College of Cardiology; ACTION, Acute Coronary Treatment and Intervention Outcomes Network; AHA, American Heart Association; DSMB, data safety monitoring
board; EP, Electrophysiology; HRS, Heart Rhythm Society; IMPROVE-HF, Improve the Use of Evidence-Based Heart Failure Therapies in the Outpatient Setting; and PI, principle investigator.

Page 95
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Appendix 2. Reviewer Relationships With Industry and Other Entities (Comprehensive)2017 AHA/ACC/HRS Guideline
for Management of Patients With Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death (July 2017)
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Reviewer Representat Employment Consultant Speakers Ownership/ Personal Institutional, Salary Expert
ion Bureau Partnership/P Research Organizational, Witness
rincipal or Other
Financial Benefit
Alfred E. Content Professor of None None None NHLBI (DSMB) Medtronic None None
Buxton Reviewer MedicineHarvard Biosense
Medical School Webster
Beth Israel
Deaconess Medical
Center
Andrew E. Content Professor of Zoll* None None Biotronik* None None Defendant,
Epstein Reviewer Medicine Boston Amiodarone
Cardiovascular Scientific* pulmonary
Division University Boston toxicity, 2016
of Pennsylvania Scientific Defendant,
Chief of Cardiology (DSMB)* Appropriaten
Section Medtronic* ess of
Philadelphia VA Medtronic pacemaker
Medical Center (DSMB) implantation,
St Jude 2016*
Medical/
Abbott*
St Jude
Medical/ Abbott
(DSMB)*
Brian Content Adjunct Professor Boehringer Lundbeck None Amarin None None Plaintiff,
Olshansky Reviewer of MedicineDes Ingelheim Inc* (DSMB)* Long QT
Moines University Lundbeck Inc* On- sudden
Professor On-X/Cryolife X/Cryolife death, 2017
Emeritus
University of Iowa
Bulent Content None None None None None None None
Gorenek Reviewer
ACC EP
Council

Page 96
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Charles I. Content Division Chief of None None None None Circulation* None None
Berul Reviewer Pediatric
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Cardiology
Children's National
Medical Center
Darren Content Executive None None None None None None None
Sudman Reviewer DirectorSimons
Fund
George J. Content Chief of Biotronik None None None None None None
Klein Reviewer CardiologyLondon Boston Scientific
Health Sciences Medtronic*
Center
Glenn N. Content Professor of None None None None None None Defendant,
Levine Reviewer MedicineBaylor Catheterizatio
ACC/AHA College of Medicine n Laboratory
Task Force DirectorCardiac Procedure,
on Clinical Care UnitMichael 2016
Practice E. DeBakey Medical Defendant,
Guidelines Center Out of
hospital
death, 2016
Gurusher S. Content Director Heart Amgen Inc. * None None None BEAT HF None None
Panjrath Reviewer Failure and ENDEAVOUR
ACC Heart Mechanical Support
Failure and ProgramGeorge
Transplant Washington
Council University

Page 97
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

James P. Official Duke University Biosense None None ARCA Biosense* ACC None
Daubert Reviewer Medical Center Webster biopharma Biotronik*
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

AHA Boston Scientific Biosense Boston


CardioFocus Webster* Scientific*
Gilead Boston Gilead Scienes,
Heart Scientific* Inc. *
Metabolics Gilead* Medtronic*
Medtronic* Gilead (DSMB) St. Jude
St. Jude Medical Medtronic* Medical*
Zoll NHLBI*
NHLBI (DSMB)
Northwestern
University
St. Jude
Medical (DSMB)
VytronUS
(DSMB)
James Content ProfessorCollege None None None AHA* ACC None Plaintiff,
Tisdale Reviewer of Pharmacy HRS* AHA Drug-induced
ACC EP Purdue University Indiana Clinical AZCert torsades de
Council Adjunct Translational QT drugs list, pointes,
ProfessorSchool Sciences credible 2017*
of Medicine Institute/Strategi meds.org
Indiana University c Research
Initiative*
John L. Sapp Official Interim Head Biosense None None Biosense ARTESiA None None
Reviewer Division of Webster* Webster* Medtronic
HRS Cardiology Medtronic Canadian Optisure
QEII Health Sciences St. Jude Institute of Registry
CentreProfessor Health Research* St. Jude
of Medicine DSMB
Dalhousie Phillips
University healthcare*
St. Jude
Medical*
Joseph Official Associate Professor None None None None UpToDate None None
Edward Reviewer of MedicineJohns
Marine ACC Hopkins University
School of Medicine

Page 98
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Kathleen T. Official Professor of None None None None None None None
Hickey Reviewer NursingColumbia
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

AHA University Medical


Center
Kenneth A. Content Chief of AHA None None AtriCure* Biosense None None
Ellenbogen Reviewer Cardiology AtriCure* Biosense Webster*
Virginia Biosense Webster* Boston
Commonwealth Webster* Boston Science*
University Medical Biotronik* Science* Circulation
Center Boston Science* Daiichi Sankyo Heart Rhythm
Capricor Medtronic* JACC
HRS Medtronic Medtronic*
Janssen (DSMB)* PACE
Medtronic* NIH* Sanofi Aventis
Pfizer* Pfizer*
Sentra heart
St. Jude
Medical*
Kim K. Content University of Jones and None None None Accreditation University of None
Birtcher Reviewer HoustonCollege Bartlett Learning Council for Houston
ACC/AHA of Pharmacology Clinical College of
Task Force Lipidology Pharmacology
on Clinical *
Practice Walgreens*
Guidelines
Kristen B. Content Duke University None None None None None None None
Campbell Reviewer Hospital
Kristen K. Content Professor of None None None None ABIM None None
Patton Reviewer Medicine ACGME
University of AHA
Washington FDA
HRS

Page 99
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

L. Brent Content Professor Boehringer None None Boston ARTESIA None None
Mitchell Reviewer Department of Ingelheim* Scientific* Health
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Cardiac Sciences Forest Protection


Libin Cardiovascular Pharmaceuticals Branch,
Institute of Alberta Guidnat Government of
University of Canada* Canada
CalgaryAlberta Medtronic
Health Services Canada*
Medtronic Inc*
Merck
Pfizer*
Servier Canada*
Martin Content I Medizinische Bayer Health None None German Centre None None None
Borggrefe Reviewer KlinikKlinikum Care for
Mannheim Boehringer Cardiovascular
GmbHUniversittskl Ingelheim Research*
inikum Impulse
Dynamics
Sanofi Aventis
St. Jude Medical
Mathew D. Official Professor of St. Jude Medical None None None None None None
Hutchinson Reviewer Medicine
HRS University of
Arizona College of
MedicineTucson
Matthew W. Content Lehigh Valley Health None None None None None None None
Martinez Reviewer Network
Sports and
Exercise EP
Council
Melissa R. Content DirectorComplex Medtronic* None None None None None None
Robinson Reviewer Ablation Program Abbott*
University of Boston
Washington Scientific*
Michael J. Content Children's Hospital None None None None None None Defendant,
Silka Reviewer Los Angeles ICD
implantation,
2017
Miguel A. Content Methodist DeBakey None None None None Houston None None
Quinones Reviewer Heart and Vascular Methodist
Center Hospital*

Page 100
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Mitchell T. Organization Jefferson Medical None None Nephroceuti None None None None
Saltzberg al CollegeChristiana cals*
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Reviewer Care Health System Stem Cell


HFSA Theranostics*
N. A. Mark Content Professor of Boston None None Boston None None None
Estes III Reviewer MedicineTufts Scientific* Scientific*
University School of Medtronic* International
Medicine St. Jude Board of Heart
Medical* Rhythm
Examiners
Medtronic*
St. Jude
Medical*
Norma M. Official New York University None None None None None None None
Keller Reviewer Medical Center
ACC
Peter Leong- Content Associate Professor Medtronic Bayer None None None Bayer None
Sit Reviewer of Medicine Canada Healthcare Healthcare
HRS Western Pharmaceutic Pharmaceutica
UniversityLondon als ls*
Health Sciences Biosense
Centre Webster
Johnson
and Johnson
Rachel J. Content Yale University Medtronic* None None Boston None None None
Lampert Reviewer School of Scientific*
MedicineSection GE Medical*
of Cardiology Medtronic, Inc.
*
St. Jude
Medical*
Sami Viskin Content Tel Aviv Medical Boston Scientific None None None None None None
Reviewer Center European Strategy
Department of Advisory Board
Cardiology
Samuel S. Content Dupont Hospital for Familial None None Familial Cardiology None None
Gidding Reviewer ChildrenNemours Hypercholesterole Hypercholestrole Division Head
ACC/AHA Cardiac Center mia Foundation mia Foundation
Task Force Regenxbio NIH Grants*
on Clinical
Practice
Guidelines

Page 101
Al-Khatib SM, et al.
2017 VA/SCD Guideline: Executive Summary

Silvia G. Content Professore Ambry Genetics None Audentes Gilead HRS None None
Priori Reviewer Ordinario di Boston Scientific Therapeutics Sciences* GS-US-372-
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Cardiologia Medtronic Inc* 1234


Universit di Medtronic, Inc.
PaviaDirettore
ScientificoIstituti
Clinici Scientifici
MaugeriPavia,
Italia
Susan Strong Official Sabin Middle School None None None None None None None
Reviewer
AHA
Win-Kuang Content Professor of None None None None None None None
Shen Reviewer Medicine
ConsultantMayo
Clinic Arizona,
Phoenix Campus
Zachary D. Official Assistant Professor RubiconMD None None None None None None
Goldberger Reviewer of Medicine
ACC/AHA Division of
Task Force Cardiology
on Clinical Harborview Medical
Practice CenterUniversity
Guidelines of Washington
Lead School of Medicine
Reviewer
This table represents the relationships of reviewers with industry and other entities that were disclosed at the time of peer review, including those not deemed to be relevant to this
document, at the time this document was under review. The table does not necessarily reflect relationships with industry at the time of publication. A person is deemed to have a significant
interest in a business if the interest represents ownership of 5% of the voting stock or share of the business entity, or ownership of $5000 of the fair market value of the business entity;
or if funds received by the person from the business entity exceed 5% of the persons gross income for the previous year. Relationships that exist with no financial benefit are also included
for the purpose of transparency. Relationships in this table are modest unless otherwise noted. Names are listed in alphabetical order within each category of review. Please refer to
http://www.acc.org/guidelines/about-guidelines-and-clinical-documents/relationships-with-industry-policy for definitions of disclosure categories or additional information about the
ACC/AHA Disclosure Policy for Writing Committees.

*Significant relationship.
No financial benefit.
This disclosure was entered under the Clinical Trial Enroller category in the ACCs disclosure system. To appear in this category, the author acknowledges that there is no direct or
institutional relationship with the trial sponsor as defined in the ACC/AHA Disclosure Policy for Writing Committees.

ACC indicates American College of Cardiology; AHA, American Heart Association; CPVT, catecholaminergic polymorphic ventricular tachycardia; DSMB, data safety monitoring board; EP,
electrophysiology; HFSA, Heart Failure Society of America; HRS, Heart Rhythm Society; ICD, implantable cardioverter-defibrillator; NHLBI, National Heart, Lung, and Blood Institute; and NIH,
National Institutes of Health.

Page 102
2017 AHA/ACC/HRS Guideline for Management of Patients With Ventricular Arrhythmias
and the Prevention of Sudden Cardiac Death: Executive Summary: A Report of the American
College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines
and the Heart Rhythm Society
Sana M. Al-Khatib, William G. Stevenson, Michael J. Ackerman, William J. Bryant, David J.
Callans, Anne B. Curtis, Barbara J. Deal, Timm Dickfeld, Michael E. Field, Gregg C. Fonarow, Anne
M. Gillis, Mark A. Hlatky, Christopher B. Granger, Stephen C. Hammill, Jos A. Joglar, G. Neal Kay,
Downloaded from http://circ.ahajournals.org/ by guest on November 14, 2017

Daniel D. Matlock, Robert J. Myerburg and Richard L. Page

Circulation. published online October 30, 2017;


Circulation is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright 2017 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7322. Online ISSN: 1524-4539

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circ.ahajournals.org/content/early/2017/10/30/CIR.0000000000000548.citation

Data Supplement (unedited) at:


http://circ.ahajournals.org/content/suppl/2017/10/30/CIR.0000000000000548.DC1
http://circ.ahajournals.org/content/suppl/2017/10/30/CIR.0000000000000548.DC2

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in
Circulation can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office.
Once the online version of the published article for which permission is being requested is located, click Request
Permissions in the middle column of the Web page under Services. Further information about this process is
available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation is online at:


http://circ.ahajournals.org//subscriptions/
Author Relationships With Industry and Other Entities (Comprehensive)2017 AHA/ACC/HRS Guideline for Management of Patients With Ventricular
Arrhythmias and the Prevention of Sudden Cardiac Death (January 2016)

Committee Employment Consultant Speakers Ownership/ Personal Research Institutional, Expert Witness
Member Bureau Partnership/ Organizational, or
Principal Other Financial Benefit
Sana M. Al-Khatib Duke Clinical Research None None None AHRQ* HRS (Board of Third Party,
(Chair) Institute; Duke University FDA* Trustees) Implantable
Professor of Medicine NHLBI* Cardioverter
PCORI* Defibrilator,
VA (DSMB) 2017
William G. Vanderbilt University St. Jude Medical Boston Biosense None Circulation (Editor) * None
Stevenson Medical CenterProfessor Scientific Webster NIH CABANA trial
(Vice Chair) OF Medicine; Brigham and VANISH trial Steering
Womens HospitalDirector Committee (Canadian
of Clinical Cardiac EP Institutes for Health
Research)
Michael J. Mayo ClinicProfessor of Boston Scientific None Transgenomic NIH* Transgenomic Defendant,
Ackerman Medicine, Pediatrics, and Gilead Sciences (Familion) * (Familion) * Long QT
Pharmacology; Long QT Invitae Blue Ox Blue Ox Health Related
Syndrome/Genetic Heart Medtronic Health Corporation Death, 2016
Rhythm Clinic and the Mayo Myokardia Corporation AliveCor
Clinic Windland Smith Rice St. Jude Medical AliveCor StemoniX
Sudden Death Genomics SADS* StemoniX
LaboratoryDirector Audentes
Therapeutics
William J. Bryant Dominick Feld Hyde None None None None Alliance for a None
Attorney at Law Healthier
Generation
AHA Corporate
Relations Review
Committee

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society.
David J. Callans University of Pennsylvania Biosense None None Biosense Webster None None
Health SystemProfessor of Webster* (PI)
Medicine; Associate Director Biotronik Endosense (PI)
of EP Boston Scientific* St. Jude Medical
Medtronic (DSMB)
St. Jude Medical Hanssen (DSMB)
nContact (DSMB)
Impulse Dynamics
(DSMB)
Anne B. Curtis University at Buffalo SUNY ACC None None NHLBI (DSMB) None None
Distinguished Professor; AHA Medtronic
Charles and Mary Bauer Daiichi-Sankyo*
Professor and Chair Medtronic*
Sanofi Aventis
Novartis
Medscape*
St. Jude Medical*
WebMD
Barbara J. Deal Getz Professor of Cardiology None None None None None None
Feinberg School of Medicine
Northwestern University
Timm Dickfeld University of Maryland Biosense None None Biosense (PI)* Impulse Dynamics Plaintiff,
Professor of Medicine Abbott/Topera* General Electric Siemens* Perforation,
St. Jude Medical (PI)* 2015
Siemens* Impulse Dynamics Plaintiff, SCD,
(DSMB) 2015
NIH
Anne M. Gillis University of Calgary AHA None None Medtronic* None Defendant,
Professor of Medicine Libin Syncope and
Cardiovascular pacemaker,
institute 2017

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society.
Christopher B. Duke Clinical Research Abbie None None Armetheon* GE Healthcare* None
Granger Institute; Duke University Armetheon AstraZeneca* Medtronic*
Professor of Medicine, AstraZeneca* Bayer* ZOLL Medical*
Cardiac Care Unit Bayer* Boehringer Spacelabs*
Boehringer Ingelheim* Phillips*
Ingelheim* Bristol-Myers
Boston Scientific Squibb*
Bristol-Myers Daiichi-Sankyo*
Squibb* FDA*
Daiichi-Sankyo* GlaxoSmithKline*
Eli Lilly* Janssen
Gilead Sciences* Pharmaceuticals*
GlaxoSmithKline* Medtronic*
Hoffman- Novartis*
LaRoche* Pfizer*
Janssen Sanofi-aventis*
Pharmaceuticals* Takeda
Medtronic* Pharmaceutical*
Medscape The Medicines
Merck Company*
Novartis*
NIH*
Pfizer*
Sanofi-aventis*
Sirtex
Takeda
Pharmaceutical*
The Medicines
Company*
Verseon*
Mark A. Hlatky Stanford University School of ACC* None None HeartFlow* None None
MedicineProfessor of Acumen* Sanofi-aventis
Health and Research Policy, Blue Cross/Blue George Institute
and of Cardiovascular Shield NHLBI (DSMB)
Medicine Genetech

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society.
Stephen C. Mayo ClinicProfessor None None None None None None
Hammill Emeritus of Medicine
Jos A. Joglar UT Southwestern Medical None None None None None None
CenterProfessor of
Internal Medicine; Clinical
Cardiac EPFellowship
Program Director
G. Neal Kay University of Alabama at None None None None None None
BirminghamProfessor
Emeritus
Michael E. Field University of Wisconsin None None None None None None
School of Medicine and
Public HealthDirector,
Clinical EP and Cardiac
Arrhythmia Service,
Associate Professor of
Medicine

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society.
Gregg C. Fonarow Ahmanson-UCLA Amgen None None Medtronic ACC/AHA Task Force None
Cardiomyopathy Center Janssen IMPROVE-HF on Data Standards
Director; UCLA Division of Pharmaceuticals (Steering ACC/AHA Task Force
CardiologyCo-Chief Medtronic Committee) on Performance
Novartis* Medtronic* Measures (Chair)
St. Jude Medical NHLBI* ACTION Registry
ZS Pharma NIH/NIAID* Research and
Novartis* Publications
Committee
AHA Workplace
Health Steering
Committee (Chair)
AHA Consumer Health
Quality Coordinating
Committee
AHA Manuscript
Oversight
Committee
GWTG Steering
Committee (PRT)
JAMA Cardiology
(Associate Editor)
Daniel D. Matlock University of Colorado None None None AFAR* ACC None
School of Medicine NIH* Circulation
Associate Professor of PCORI* Cardiovascular
Medicine
Quality and
Outcomes
Medical Decision
Making
Journal of Palliative
Medicine
Robert J. Myerburg University of Miami Miller None None None Miami Heart None Defendant,
School of Medicine Research Various
Professor of Medicine and Foundation Medical
Physiology VEST (DSMB) Cases, 2015*

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society.
Richard L. Page University of Wisconsin None None None None FDA None
Hospital & ClinicsChair,
Department of Medicine

This table represents all relationships of committee members with industry and other entities that were reported by authors, including those not deemed to be relevant to this
document, at the time this document was under development. The table does not necessarily reflect relationships with industry at the time of publication. A person is deemed
to have a significant interest in a business if the interest represents ownership of 5% of the voting stock or share of the business entity, or ownership of $5,000 of the fair
market value of the business entity; or if funds received by the person from the business entity exceed 5% of the persons gross income for the previous year. Relationships that
exist with no financial benefit are also included for the purpose of transparency. Relationships in this table are modest unless otherwise noted. Please refer to
http://www.acc.org/guidelines/about-guidelines-and-clinical-documents/relationships-with-industry-policy for definitions of disclosure categories or additional information
about the ACC/AHA Disclosure Policy for Writing Committees.

*Significant relationship.
No financial benefit.
This disclosure was entered under the Clinical Trial Enroller category in the ACCs disclosure system. To appear in this category, the author acknowledges that there is no direct
or institutional relationship with the trial sponsor as defined in the ACC/AHA Disclosure Policy for Writing Committees.

ACC indicates American College of Cardiology; ACTION, Acute Coronary Treatment and Intervention Outcomes Network; AHA, American Heart Association; AFAR, American
Federation for Aging Research; CABANA; Catheter Ablation Versus Antiarrhythmic Drug Therapy for Atrial Fibrillation; DSMB, data safety monitoring board; EP,
Electrophysiology; GWTG, Get With The Guidelines; FDA, Food and Drug Administration; HRS, Heart Rhythm Society; IMPROVE-HF, Registry to Improve the use of Evidence-
Based Heart Failure Therapies in the Outpatient Setting; JAMA, Journal of the American Medical Association; NIH, National Institutes of Health; NHLBI, National Heart, Lung,
and Blood Institute; NIAID, National Institute of Allergy and Infectious Diseases; PACES, Pediatric and Congenital Electrophysiology Society; PCORI, Patient Centered Outcomes
Research Institute; PI, principle investigator; PRT, pharmaceutical round table; SADS, Sudden Arrhythmia Death Syndromes Foundation; SMDM, Society for Medical Decision
Making; UK, United Kingdom; and VANISH, Vasopressin Versus Noradrenaline as Initial Therapy in Septic Shock.

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society.
2017 VA/SCD Guideline Data Supplement
Table of Contents
Data Supplement 1. Nonrandomized Trials, Observational Studies, and/or Registries for History and Physical Examination (Section 4.1) ................................. 6
Data Supplement 2. Nonrandomized Trials, Observational Studies, and/or Registries of Noninvasive Evaluation (12-lead ECG, Exercise Testing and
Electrocardiographic Monitoring) (Section 4.2.1) ............................................................................................................................................................................ 8
Data Supplement 3. RCTs Comparing Ambulatory Electrocardiography (Section 4.2.2) ............................................................................................................... 14
Data Supplement 4. Nonrandomized Trials, Observational Studies, and/or Registries of Ambulatory Monitors (Section 4.2.2) ................................................ 15
Data Supplement 5. Nonrandomized Trials, Observational Studies, and/or Registries of Implanted Cardiac Monitors (Section 4.2.3) ...................................... 16
Data Supplement 6. Nonrandomized Trials, Observational Studies, and/or Registries Comparing Noninvasive Cardiac Assessment (Section 4.2.4) ................. 18
Data Supplement 7. Nonrandomized Trials, Observational Studies, and/or Registries Comparing Biomarkers (Section 4.2.5)................................................... 19
Data Supplement 8. RCTs Evaluating EP Study for VA (Section 4.3.2) ........................................................................................................................................... 22
Data Supplement 9. Nonrandomized Trials, Observational Studies, and/or Registries of EP Study for VA - (Section 4.3.2) ........................................................... 26
Data Supplement 10. RCTs for Preventing SCD with HF Medications - (Section 5.2) ....................................................................................................................... 32
Data Supplement 11. RCTs and Nonrandomized Trials, Observational Studies, and/or Registries Related to Surgery and Revascularization Procedures (Section
5.5) .................................................................................................................................................................................................................................................... 37
Data Supplement 12. Nonrandomized Trials, Observational Studies, and/or Registries of Arrhythmic Surgery and Revascularization for Arrhythmia
Management (Section 5.5.1) ......................................................................................................................................................................................................... 44
Data Supplement 13. RCTs for Autonomic Modulation (Section 5.6)............................................................................................................................................ 48
Data Supplement 14. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries for Autonomic Modulation (Section 5.6) .......................... 50
Data Supplement 15. RCTs Comparing Acute Management of Specific Arrythmias - (Section 6) .................................................................................................... 51
Data Supplement 16. Nonrandomized Trials, Observational Studies, and/or Registries Comparing Acute Management of Specific Arrythmias (Section 6) ..... 58
Data Supplement 17. RCTs Secondary Prevention Sudden Death in Ischemic Heart Disease (Section 7.1.1)............................................................................... 64
Data Supplement 18. Nonrandomized Trials, Observational Studies, and/or Registries for Secondary Prevention Sudden Death in Ischemic Heart Disease
(Section 7.1.1) ................................................................................................................................................................................................................................... 69
Data Supplement 19. Nonrandomized Trials, Observational Studies, and/or Registries for Coronary Artery Spasm (Section 7.1.1.1) ....................................... 71
Data Supplement 20. Nonrandomized Trials, Observational Studies, and/or Registries for Post CABG VT/VF (Section 7.1.1.2) ................................................. 75
Data Supplement 21. RCTs and Nonrandomized Trials, Observational Studies, and/or Registries of ICDs Primary Prevention VA and Sudden Death in Patients
with Ischemic Cardiomyopathy (Section 7.1.2) ............................................................................................................................................................................. 77
Data Supplement 22. RCTs Evaluating Treatment and Prevention of Recurrent VA in Patients with Ischemic Heart Disease (Section 7.1.3) ............................. 84
Data Supplement 23. Nonrandomized Trials, Observational Studies, and/or Registries of Treatment of Recurrent Arrhythmias in IHD (Section 7.1.3) ........... 90
Data Supplement 24. Nonrandomized Trials, Observational Studies, and/or Registries of NICM (Section 7.2) ........................................................................... 98
Data Supplement 25. RCTs Secondary Prevention SCD in NICM (Section 7.2.1) ......................................................................................................................... 107
Data Supplement 26. Nonrandomized Trials, Observational Studies, and/or Registries of Secondary Prevention SCD in NICM (Section 7.2.1) ....................... 112
Data Supplement 27. RCTs Primary Prevention SCD in NICM (Section 7.2.2) ............................................................................................................................. 116
Data Supplement 28. Nonrandomized Trials, Observational Studies, and/or Registries of Primary Prevention of SCD in NICM (Section 7.2.2) ....................... 119

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 29. RCTs and Nonrandomized Trials, Observational Studies, and/or Registries of Treatment of Recurrent VA in Patients With NICM (Section
7.2.3) ............................................................................................................................................................................................................................................... 125
Data Supplement 30. Nonrandomized Trials, Observational Studies, and/or Registries of Arrhythmogenic Right Ventricular Cardiomyopathy (Section 7.3) 127
Data Supplement 31. Nonrandomized Trials, Observational Studies, and/or Registries of Hypertrophic Cardiomyopathy (Section 7.4) ................................. 145
Data Supplement 32. Nonrandomized Trials, Observational Studies, and/or Registries of Myocarditis (Section 7.5) ............................................................... 159
Data Supplement 33. Nonrandomized Trials, Observational Studies, and/or Registries of Cardiac Sarcoidosis (Section 7.6) ................................................... 160
Data Supplement 34. Nonrandomized Trials, Observational Studies, and/or Registries of Other Infiltrative Cardiomyopathies (Section 7.6.1) ...................... 168
Data Supplement 35. Nonrandomized Trials, Observational Studies, and/or Registries of Use of ICD and WCD in Patients with HFrEF - (Section 7.8.1) ........... 170
Data Supplement 36. Nonrandomized Trials, Observational Studies, and/or Registries Related to LVAD (Section 7.8.3) ......................................................... 173
Data Supplement 37. Nonrandomized Trials, Observational Studies, and/or Registries Related to ICD Use After Heart Transplantation (Section 7.8.4) ........ 173
Data Supplement 38. Nonrandomized Trials, Observational Studies, and/or Registries Evaluating the Risk of Sudden Death or VA in Patients with
Neuromuscular Disorders (Section 7.8) ....................................................................................................................................................................................... 175
Data Supplement 39. Nonrandomized Trials Related to Cardiac Channelopathies (Section 7.9) ............................................................................................... 184
Data Supplement 40. Nonrandomized Trials Related to Congenital LQTS (Section 7.9.1.1.) ...................................................................................................... 196
Data Supplement 41. Nonrandomized Trials Related to Catecholaminergic Polymorphic Ventricular Tachycardia (Section 7.9.1.2.) ...................................... 207
Data Supplement 42. Nonrandomized Trials Related to Brugada Syndrome (Secction7.9.1.3) .................................................................................................. 211
Data Supplement 43. Nonrandomized Trials Related to Early Repolarization J-wave Syndrome (Secction 7.9.1.4) ............................................................... 221
Data Supplement 44. Nonrandomized Trials Related to Short-QT Syndrome (Secction 7.9.1.5) ................................................................................................ 225
Data Supplement 45. RCTs Related to VA in the Structurally Normal Heart (Section 8) ............................................................................................................ 228
Data Supplement 46. Nonrandomized Trials, Observational Studies, and/or Registries Related to Outflow Tract and AV Annular VA (Section 8.1) .............. 230
Data Supplement 47. Nonrandomized Trials, Observational Studies, and/or Registries of Catheter Ablation in Papillary Muscle VA - (Section 8.2) ................. 238
Data Supplement 48. Nonrandomized Trials, Observational Studies, and/or Registries Related to Interfascicular Reentrant VT (Belhassen Tachycardia)-
(Section 8.3) .................................................................................................................................................................................................................................... 241
Data Supplement 49. Nonrandomized Trials, Observational Studies, and/or Registries Related to Idiopathic Polymorphic VT/VF - (Section 8.5) ...................... 242
Data Supplement 50. Nonrandomized Trials, Observational Studies, and/or Registries of PVC-induced Cardiomyopathy - (Section 9) ...................................... 247
Data Supplement 51. Nonrandomized Trials, Observational Studies, and/or Registries Related to Pregnancy - (Section 10.2) ................................................... 258
Data Supplement 52. RCTs Comparing Medication-Induced Arrhythmias - (Section 10.7)............................................................................................................ 267
Data Supplement 53. Nonrandomized Trials, Observational Studies, and/or Registries of Medication-Induced Arrhythmias (Section 10.7) .............................. 268
Data Supplement 54. Nonrandomized Trials, Observational Studies, and/or Registries Related to ACHD - (Section 10.8) .......................................................... 274
Data Supplement 55. Nonrandomized Trials, Observational Studies, and/or Registries of S-ICD - (Section 11.1) ........................................................................ 293
Data Supplement 56. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries for WCD (Section 11.2) .................................................... 299
Data Supplement 57. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries for Special Considerations for Catheter Ablation (Section
12) ................................................................................................................................................................................................................................................... 300
Data Supplement 58. Nonrandomized Trials, Observational Studies, and/or Registries Related to Post-Mortem Evaluation of SCD - (Section 13) ................... 304
Data Supplement 59. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries of Terminal Care - (Section 14)........................................... 308
Data Supplement 60. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries for Shared Decision Making (Section 15) ........................ 314
Data Supplement 61. Randomized Trials, Observational Studies, and/or Registries Related to Cost and Value Considerations - (Section 16)............................ 317

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
References: ..................................................................................................................................................................................................................................... 321

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Methodology and Evidence Review
The recommendations listed in this guideline are, whenever possible, evidence based. An extensive evidence review was conducted from April through
September 2016, that included literature published through September 2016. Other selected references published through March 2017 were incorporated by
the writing committee. Literature included was derived from research involving human subjects, published in English, and indexed in MEDLINE (through
PubMed), EMBASE, the Cochrane Library, the Agency for Healthcare Research and Quality, and other selected databases relevant to this guideline. Key search
words included but were not limited to the following: accelerated idioventricular rhythm, advanced cardiac life support, ambulatory electrocardiography,
amiodarone, amyloidosis, Antiarrhythmic drugs ARNI Angiotensin Receptor-Neprilysin Inhibitor, arrhythmias, arrhythmogenic right ventricular dysplasia,
atenolol, autonomic modulation, biomarkers, CABG, cardiac, catheter ablation, cardiac arrest, cardiac arrhythmia, cardiac catheterization, cardiac magnetic
resonance imaging, cardiac sympathetic denervation, cardiac troponin, cardiomyopathy, catecholaminergic polymorphic ventricular tachycardia, carvedilol,
choice behavior, coronary artery bypass surgery, coronary stent, cryoablation deactivation, decision-making, digoxin toxicity, dilated cardiomyopathy, dilated
non ischemic cardiomyopathy, disease management, Dor Procedure, drug induced arrhythmia, drug induced long QT, emergency medical services, electrical
storm, electrocardiography, electrophysiologic study, electrophysiologic techniques, electrophysiological testing, emergency management, end of life,
endocardiectomy exercise test, Fabrys disease, fibrillation, flecainide,heart arrest, heart disease, hemochromatosis, hemodynamically stable ventricular
tachycardia, holter monitor, hypertrophic, implantable cardiac monitor, incessant, infiltrative heart disease, intervention, lamin a/c left ventricular assist
device, left ventricular reconstruction, lidocaine, long QT syndrome, loop recorder, LV dysfunction, metoprolol, monomorphic, muscular dystrophies,
myocardial infarction/therapy, myotonic dystrophy, nadolol, natriuetic peptides, papillary muscle, patient perspective, patient preference, percutaneous
coronary, polymorphic, Polymorphous Ventricular Tachycardia, premature ventricular contractions, procainamide, propranolol, pulseless electrical activity,
PVC induced cardiomyopathy, resting ecg, renal denervation, resuscitation, risk stratification, secondary prevention, shared decision making, sotalol, spinal
cord stimulation, subcutaneous implantable cardioverter defibrillators, sudden cardiac death, sudden death, syncope, tachycardia, torsades de pointes, vagal
nerve stimulation ventricular, ventricular arrhythmias, ventricle extrasystole, ventricular fibrillation, ventricular premature complexes, ventricular tachycardia

Abbreviations: 1 indicates primary; 2, secondary; AAD, antiarrhythmic drugs; ACA, aborted cardiac arrest; ACC, American College of Cardiology; ACHD, adult
congenital heart disease; ACLS, advanced cardiac life support; ACS, acute coronary syndrome; AF, atrial fibrillation; AHA, American Heart Association; AMI,
acute myocardial infarction; ARVC, arrhythmogenic right ventricular cardiomyopathy; AS, atrial stenosis; AT, atrial tachyarrhythmias; AV, atrioventricular;
AVID, antiarrhythmics versus implantable defibrillators; BB, beta blocker; BBB, bundle branch block; BBRVT, bundle branch reentrant ventricular tachycardia;
BID, two times a day; BNP, brain natriuretic peptide; BP, blood pressure; BrS, Brugada syndrome; CA, cardiac arrest; CABG, coronary artery bypass graft;
CABG-PATCH, coronary artery bypass graft patch trial; CAD, coronary artery disease; CASH, cardiac arrest study Hamburg; CASS, coronary artery surgery
study; CE, cardiac event; CHF, congestive heart failure; CHFSTAT, survival trial of antiarrhythmic therapy in congestive heart failure; CI, confidence interval;
CIBIS II, cardiac insufficiency bisoprolol study II; CIDS, Canadian implantable defibrillator; ICD, cardiovascular implantable electronic device; CMRI, cardiac
magnetic resonance imaging; COPD, chronic obstructive pulmonary disease; CPR, cardiopulmonary resuscitation; CPVT, catecholaminergic polymorphic
ventricular tachycardia; CRT, cardiac reshynchronization therapy; CS, carotid sarcoidosis; CT, computed tomography; CV, cardiovascular; CVD, cardiovascular
disease; CXR, chest x-ray; DCM, dilated cardiomyopathy; DEFINITE, defibrillator in nonischemic cardiomyopathy treatment evaluation; DFT, defibrillation
threshold; DINAMIT, defibrillator in acute myocardial infarction trial; DM1, myotonic dystrophy 1; DM2, myotonic dystrophy; DYS, dystrophin;ECG,
electrocardiogram; EDMD2, Emery-Dreifuss muscular dystrophy type 2; EF, ejection fraction; EFFORTLESS S-ICD, evaluation of factors impacting clinical
outcome and cost effectiveness of the S-ICD; EGM, electorgram EMD, electromechanical dissociation; EP, electrophysiological; EPS, electrophysiological study;
ERP, effective refractory period; ESRD, end stage renal disease; EURO-VT Study, Euroventricular tachycardia study; GDMT, guideline-directed
management and therapy; GFR, glomerular filtration rate; HCM, hypertrophic cardiomyopathy; HELP-VT, heart center of Leipzig VT study; HF, heart failure;

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
HPS, His-Purkinje system; HR, hazard ratio; HTN, hypertension; Hx, history; HV, His Purkinje conduction rate; ICD, implantable cardioverter-defibrillator; ICU,
intensive care unit; IDCM, idiopathic dilated cardiomyopathy; IDE, investigational device exemption; ILR, implantable loop recorder; IRIS, insulin resistance
intervention after stroke; IV, intravenous; KM, Kaplan-Meier; LBBB, left bundle branch block; LCSD, left cardiac sympathetic denervation; LGE, late gadolinium
enhancement; LQTS, long QT syndrome; LV, left ventricle; LVAD, left ventricular assist device; LVEF, left ventricular ejection fraction; LVH, left ventricular
hypertrophy; LVOT, left ventricular outflow tract; MACE, major adverse cardiac event; MADIT, multicenter automatic defibrillator implantation trial; MAGIC,
magnesium in coronaries; MD, muscular dystrophy; MI, myocardial infarction; MR, mitral regurgitation; MRI, magnetic resonance imaging; MTWA, microvolt
T-wave alternans; MUSTT, multicenter unsustained tachycardia trial; N/A, not available; NICM, nonischemic dilated cardiomyopathy; NPV, negative predictive
value; NS, not significant; NSVT, nonsustained ventricular tachycardia; NYHA, New York Heart Association classification for heart failure; NT-proBNP, N-
terminal pro b-type natriuretic peptide; OHCA, out-of-hospital cardiac arrest; OPTIC, optimal pharmacological therapy in cardioverter defibrillator patients;
OR, odds ratio; PainFREE Rx II, pacing fast ventricular tachycardia reduces shock therapies; PARADIGM-HF, prospective comparison of ARNI with ACEI to
determine impact on global mortality and morbidity in heart failure trial; PCI, percutaneous coronary intervention; PE, physical examination; PES,
programmed electrical stimulation; PM, papillary muscle; PMCD, Perimortem Cesarian Delivery; PMCS, Perimortem Cesarian Section; PMVT, polymorphic
ventricular tachycardia; PO, per os; PROCAT, Parisian region out of hospital cardiac arrest; PVC, premature ventricular contractions; PVR, pulmonary valve
replacement; QoL, quality of life; RBB, right bundle branch; RBBB, right bundle branch block; RCSD right cardiac sympathetic denervation; RCT, randomized
controlled trials; RNA, radionuclide angiography; RR, relative risk; RRR relative risk ratio; RyR2, Ryanodine receptor type 2; S-ICD, subcutaneous implantable
cardioverter-defibrillator; SAECG, signal averaged ECG; SBP, systolic blood pressure; SCA, sudden cardiac arrest; SCD, sudden cardiac death; SCD-HeFT, sudden
cardiac death in heart failure trial; SCS, spinal cord stimulation; SHD, structural heart disease; SMASH VT, substrate mapping and ablation in sinus rhythm to
halt ventricular tachycardia; SND, sinus node dysfunction; SQTS, short QT syndrome; STICH, surgical treatment for ischemic heart failure; STICHES, surgical
treatment for ischemic heart failure extension study; SVT, supraventricular tachycardia; SYNTAX, synergy between PCI with Taxus and cardiac surgery; TdP,
torsades de pointes; TIA, transient ischemic attack; TOF, tetralogy of Fallot; VA, ventricular arrhythmias; VALIANT, valsartan in acute myocardial infarction;
VANISH, ventricular tachycardia ablation versus escalated antiarrhythmic drug therapy in ischemic heart disease; VERP, ventricular effective refractory period;
VF, ventricular fibrillation; VFL, ventricular flutter; VHD, valvular heart disease; VT, ventricular tachycardia; VTE, ventricular tachyarrhythmic events; and WCD,
wearable cardiac defibrillator.

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 1. Nonrandomized Trials, Observational Studies, and/or Registries for History and Physical Examination (Section 4.1)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Ruwald, et al. 2012 Study type: Inclusion criteria: 1 endpoint: The incidence rates observed are
(1) Retrospective Patients hospitalized or Incidence of syncope and higher than previously reported
22588456 observational study seen in emergency associations with comorbidities and and the age distribution of syncope
from a registry cohort department with first pharmacotherapy is widely different according to
with matched episode of syncope Results: gender. Syncope is more common
controls. between 1997 and 2009. Age distribution peaked at 20, 60, in females, in the elderly, is
Exclusion criteria: and 80 y. Incidence was higher in generally a diagnosis associated
Size: 127,508 patients Not specified women in all age groups, although with considerable comorbidity.
with first episode of the peak in the oldest age group The data may be influenced by
syncope. Each subject occurred 57 y earlier in men. CVD the fact that the study is
paired with 5 age and was present in 28% of the subjects, dominated by syncope leading to
sex matched controls. and drug therapy was being used by hospitalization and emergency
48%. There was an association department visits.
between CVD and admission for
syncope, inversely related to age -
029 y (OR: 5.8); 3049 y (OR: 4.4);
5079 y (OR: 2.9), and >80 y (OR:
2.0). Cardiovascular
pharmacotherapy associated with
age and risk of syncope was similar.
Soteriades et al. Study type: Inclusion criteria: 1 endpoint: Cardiac syncope constitutes a
2002 (2) Retrospective analysis Reported episodes of Death from any cause, MI or death high-risk group for morbidity and
12239256 of a prospectively syncope by subjects in from coronary heart disease, and premature mortality from CVD.
enrolled long term Framingham study fatal or nonfatal stroke. Patients with unknown cause are
population cohort population examined Results: Overall incidence of a first a mixed group at apparent
(Framingham) between 1971 and 1998. report of syncope was 6.2 per 1000 increased risk for death and
Reports coded as yes, person-y, with an increase with warrant further diagnostic testing.
Size: 727 patients with no, or maybe. increasing age, most prominent at Vasovagal syncope has a benign
reported syncope and 70 y. Age-adjusted incidence was 7.2 prognosis.
long term follow up Exclusion criteria: per 1000 person-y among both men
from a population of Equivocal reports of and women. Causes among men and
7814 participants syncope (N=120), women were: cardiac causes (13.2%
(3563 men and 4251 participants who had not and 6.7%), unknown (31.0% 40.7%),

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
women) followed for had an examination stroke or TIA (4.3%and 4.0%),
an average of 17 y in within 4 y of the report seizure disorder (7.2% and 3.2%),
the outcome analysis. (N=101), syncope due to vasovagal (19.8% 22.2%), orthostatic
head trauma (N=47), (8.6% and 9.9%), medication (6.3%
incomplete records and 7.2%), and other (9.5% and
(N=7). 6.1%). Recurrences were reported in
21.6%).
There were 847 deaths from all
causes, 263 MI or deaths from
coronary heart disease, and 178
fatal or nonfatal strokes during a
mean follow-up of 8.6 y (median,
7.7). Participants with cardiac
syncope had lower survival than
those without syncope.
Middlekauff et al. Study type: Inclusion criteria: 1 endpoint: Patients with advanced HF and
1993 (3) Retrospective analysis Consecutive series of SCD syncope are at increased risk of all
8417050 of a consecutive patients with advanced cause mortality, largely associated
patient cohort HF without a Hx of CA Results: After a mean follow-up with an increased risk of SCD.
referred for optimization of 365+419 d, 165 patients (35%)
Size: 491 patients of medical therapy, often were alive, 148 (30%) had
in conjunction with pre- undergone heart transplantation, 69
transplant evaluation, (14%) had died suddenly, 66 (13%)
between 1983 and 1991 had died of progressive HF, 19 (4%)
had died of noncardiac or unknown
Exclusion criteria: causes and 24 (4%) were lost to
Prior Hx of CA. follow-up. All-causes at I y
was 29% and sudden death was
15%. All cause mortality was greater
in patients with syncope
(65% vs. 25%, p<0.00001). SCD risk
was significantly greater in patients
with syncope (45% vs. 12%, p
<0.00001).

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 2. Nonrandomized Trials, Observational Studies, and/or Registries of Noninvasive Evaluation (12-lead ECG, Exercise Testing
and Electrocardiographic Monitoring) (Section 4.2.1)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Steinman et al. Study type: retrospective Inclusion criteria: regular 1 endpoint: diagnosis of VT VT is the most common diagnosis
1989 (4) cohort wide QRS tachycardia in in adults with stable, wide complex
2915409 conscious adults Results: 75% of patients had tachycardia
Size: 20 patients atherosclerotic heart disease, with
Exclusion criteria: remote MI in 73%
hemodynamic instability Diagnosis of VT established in
17/20 patients, by AV dissociation
or the use of Wellenscriteria. EP
testing in 17 patients confirmed
the diagnosis of VT in 94%.

Brugada et al. Study type: prospective Inclusion criteria: ECGs 1o endpoint: mechanism Absence of RS in all precordial
1991 (5) cohort with wide QRS (>0.12 s) confirmed by EPS leads was highly specific for VT
2022022 When RS is present in 1 or more
Size: 554 tachycardias Exclusion criteria: AAD Results: New criteria had precordial leads, RS interval of >100
treatment sensitivity of 0.987 and specificity ms is highly specific for VT
of 0.965. Other criteria included AV
dissociation and morphology in leads
V1-2 and V6
Wellens HJ et Study type: Prospective Inclusion criteria: 1o endpoint: development of Capture or fusion beats seen only
al. 1978 (6) cohort Diagnosis confirmed by algorithm for differentiation of VT infrequently
623134 His bundle ECG recording from SVT
Size: 140 ECGs, 70 of
sustained VT and 70 SVT Exclusion criteria: Atrial Results: Findings suggestive of VT:
with aberrancy, in 122 fibrillation or flutter in QRS >0.14 s; left axis deviation;
patients patients with SVT QRS morphology; AV dissociation
Elhendy et al. Study type: retrospective Inclusion criteria: 1 endpoint: cardiac death or 41 patients had NSVT.
2002 (7) cohort analysis intermediate pre-test nonfatal MI Study was aimed more at ischemic
12106835 probability of CAD outcomes than arrhythmias.
Size: 1460 Results: Exercise-induced VA
Exclusion criteria: Hx of occurred in 146 patients (10%).
MI or revascularization,
8

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
CAD documented on During follow-up (median 2.7 y), 1
angiography, or LBBB endpoint occurred in 36 patients.
In multivariate analysis,
independent predictors of cardiac
events were exercise-induced VA
(chi-square 4.7, p=0.03) and
exercise heart rate (chi-square 18,
p=0.0001).
Grady et al. Study type: retrospective Inclusion criteria: 1 endpoint: All-cause mortality, Exercise-induced LBBB predicts a
1998 (8) matched control cohort Exercise-induced LBBB PCI, open heart surgery, nonfatal higher risk of death and major
9440667 study MI, documented symptomatic or cardiac events.
Exclusion criteria: sustained VT, or implantation of a
Size: 70 cases and 70 preexcitation or permanent pacemaker or an ICD.
matched controls permanent pacemakers Results: 37 events (28 in LBBB, 9
in controls) occurred during mean
3.7 y follow-up
Adjusted relative risk in LBBB was
2.78 (95% CI: 1.166.65, p=0.02)
ABCD Study type: prospective, Inclusion criteria: 1 endpoint: appropriate ICD Combination of MTWA and EPS
Costantini et al. non-randomized cohort ischemic discharge or SCD identifies a subset of patients most
2009 (9) cardiomyopathy, EF<40%, likely to benefit from ICD.
19195603 Size: 566 patients and NSVT Results: 39 patients (7.5%) met Negative predictive value is not
the 1 endpoint after a median 100%, indicating that a small subset
Exclusion criteria: follow-up of 1.9 y; MTWA had a of patients may still have events even
unstable CAD, NYHA class positive predictive value of 9% and if both tests are negative.
IV HF, prior CA, sustained NPV of 95%, comparable to EPS
VA, unexplained syncope; (11% and 95% respectively)
recent (<28 d) MI, CABG, Event rate with both positive
or PCI; permanent AF; MTWA and EPS was 12%, vs. 2%
taking AAD at baseline with both negative (p=0.017)
Desai et al. Study type: retrospective Inclusion criteria: 1 endpoint: cardiovascular death 801 patients (1.8%) had a QRS>120
2006 (10) Patients with ECGs at a ms; another 2300 had BBB
16828632 Size: 46,933 consecutive single center Results: After adjustment in the No specific information on
patients with ECGs Cox model for age, gender, and arrhythmic death
Exclusion criteria: heart rate, the QRS duration score
preexcitation; BBB or was a strong independent
predictor of cardiovascular

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
paced patients mortality. For every 10ms increase
considered separately in QRS duration, there was an 18%
increase in cardiovascular risk.
Freedman et al. Study type: retrospective Inclusion criteria: All 1 endpoint: mortality Mean EF in LBBB patients 40% vs.
1987 (11) patients from CASS; BBB 49% in RBBB and 57% in patients
3597997 Size: 15,609 patients from patients compared to Results: LBBB associated with 5- without BBB
the CASS study (Coronary those without fold greater mortality; RBBB 2-fold
Artery Surgery Study); 522 greater mortality (p<0.0001 for
with BBB Exclusion criteria: both)
preexcitation, ventricular
pacing, nonspecific IVCD,
previous myocardial
surgery
Baldasseroni et Study type: retrospective Inclusion criteria: 1 endpoint: mortality LBBB Is associated with higher
al. 2002 (12) analysis of outpatient unselected outpatients mortality in CHF
11868043 registry with HF Results: LBBB was present in 1391
patients (25.2%) and was
Size: 5517 patients Exclusion criteria: N/A associated with an increased 1y
mortality rate from any cause (HR
1.70; 95% CI: 1.412.05) and
sudden death (HR: 1.58; 95% CI:
1.212.06).

MUSTT Study type: retrospective Inclusion criteria: CAD, 1 endpoint: CA or arrhythmic Likely reflects the effect of
Zimetbaum et substudy EF<40%, NSVT death ventricular dyssynchrony
al. 2004 (13)
15289365 Size: 431 Exclusion criteria: Results: LBBB and intraventricular
treatment with AAD or an conduction delay were associated
ICD with a 50% increase in the risk of
both arrhythmic and total
mortality. RBBB was not
associated with arrhythmic or total
mortality. LVH was the only ECG
predictor of arrhythmic (HR 1.35;
95% CI: 1.081.69) but not total
mortality.

10

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Buxton et al. Study type: retrospective Inclusion criteria: 1 endpoint: recurrence of VT/VF QRS duration is not useful in
2005 (14) substudy from PainFREE Rx patients in the study with predicting recurrent VT/VF.
16022960 II CAD and a baseline ECG. Results: QRSd was <120 ms in 291
of 431 (68%) patients (LBBB 65,
Size: 431 patients Exclusion criteria: HCM, RBBB 48, IVCD 124). Over 12mo
BrS, LQTS follow-up, VT/VF occurred in 95
(22%) patients (22% of patients
with QRSd <120ms vs.
23% of patients with QRSd
>120ms, p=NS).

MADIT-II Study type: substudy of Inclusion criteria: CAD, EF 1 endpoint: appropriate ICD Increased TWA and QTV are
Monasterio et prospective clinical trial <30% therapy and SCD independent predictors of
al. 2013 (15) appropriate ICD therapy in MADIT-II
24028998 Size: 175 patients Exclusion criteria: AF; Results: Neither QTV nor TWA patients with elevated heart rate at
heart rate <80 beats/min predicted SCD. Appropriate ICD baseline.
therapy was predicted by
combining IAA90 from T wave
alternans testing and QTVN after
adjusting for relevant correlates.

MASTER Study type: prospective, Inclusion criteria: post- 1 endpoint: SCD or appropriate Total mortality was significantly
Chow et al. non-randomized cohort MI, EF<30% ICD therapy increased in MTWA non-negative
2008 (16) study of MTWA testing patients (HR: 2.04; 95% CI: 1.10
18992649 Exclusion criteria: AF or Results: SCD or appropriate ICD 3.78; p=0.02).
Size: 575 patients; all atrial flutter, Hx of therapy occurred in 48 of MTWA did not identify patients at a
received ICDs sustained VT/VF or CA, 361 (13%, 6.3%/y) MTWA non- higher risk of a VT.
MI in past mo, negative and 22 of 214 (10%,
revascularization within 3 5.0%/y) MTWA negative patients.
mo, class IV CHF, A non-negative
advanced MTWA test result was not
cerebrovascular disease associated with 1 endpoint (HR:
1.26; 95% CI 0.762.09; p=0.37)

Gupta et al. Study type: meta-analysis Inclusion criteria: 1 endpoint: VT events were Negative MTWA result would
2012 (17) predominantly prior MI defined as the total and decrease the annualized risk of VTE
22424005 from 8.85% to 6.37% in MADIT-II

11

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 20 prospective cohort or left ventricular arrhythmic mortality and nonfatal type patients and from 5.91% to
studies consisting of 5,945 dysfunction sustained or ICD-treated VT 2.60% in SCD-HeFTtype patients.
subjects Despite a modest association,
Exclusion criteria: Results: Although there was a results of spectrally derived MTWA
healthy patients; BrS; modest association between testing do not sufficiently modify the
LQTS positive MTWA and VTE (RR: 2.45; risk of VTE to change clinical
95% CI:1.58-3.79) and nonnegative decisions
MTWA and VTE (RR: 3.68; 95% CI:
2.236.07), test performance was
poor (positive MTWA: LR+ 1.78,
LR 0.43; nonnegative MTWA: LR+
1.38, LR 0.56)
MADIT-II Study type: substudy of Inclusion criteria: prior 1 endpoint: SCD in the medically Prolonged QRS does not predict
Dhar et al. 2008 randomized clinical trial MI, EF <30% treated arm and SCD or first SCD/VT/VF in ICD treated patients
(18) that estimated the appropriate ICD therapy for rapid but does predict SCD in medically
18534364 association of prolonged Exclusion criteria: VT/VF in the ICD-treated arm treated patients.
QRSd >140ms with indicated for an ICD;
arrhythmic outcomes NYHA class IV; coronary Results: In the medically treated
revascularization arm, prolonged QRS was a
Size: 1232 patients within the preceding 3 significant independent predictor
mo; MI within the past of SCD (HR: 2.12; 95% CI1.203.76,
mo; advanced p=0.01). In the ICD-treated
cerebrovascular disease; arm, prolonged QRS did not
other potentially life- predict SCD or rapid VT/VF (HR:
threatening conditions 0.77; 95% CI 0.471.24, p=0.28).

Bloomfield et Study type: prospective Inclusion criteria: prior 1 endpoint: 2y all-cause mortality Among MADIT IIlike patients,
al. 2004 (19) cohort MI, EF<30% MTWA is better than QRS duration at
15451804 Results: For abnormal MTWA identifying a high-risk group; it is also
Size: 177 patients Exclusion criteria: AF or compared to normal (negative) better at identifying a low-risk group
atrial flutter; requirement test, the HR: 4.8; p=0.02; for QRS unlikely to benefit from ICD therapy.
for >120ms compared to <120ms, the
ventricular pacing; HR for 2y mortality was 1.5
unstable CAD; NYHA class (p=0.367). The actuarial mortality
IV HF; unable to exercise rate was substantially lower
on a bicycle or treadmill among patients with normal
MTWA (3.8%; 95% CI: 09.0) than

12

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
the mortality rate in patients with
a narrow QRS (12.0%; 95% CI: 5.6
18.5).
Iuliano et al. Study type: retrospective Inclusion criteria: 1 endpoint: total mortality and QRS prolongation is an
2002 (20) analysis of CHF-STAT ischemic or nonischemic sudden death independent predictor of both
12075267 cardiomyopathy, NYHA increased total mortality and sudden
Size: 669 patients class II-IV, >10 PVCs/h, EF Results: Prolonged QRS (>120 ms) death in patients with HF.
<40% was associated with a significant
increase in mortality (49.3% vs
Exclusion criteria: recent 34.0%, p=0.0001) and sudden
MI, Hx of ACA, QRS death (24.8% vs 17.4%, p=0.0004).
>180ms, LBBB was associated with worse
or a QTc >500ms survival (p=0.006) but not sudden
death
Perez-Rodon, et Study type: Retrospective Inclusion criteria: 1 endpoint: all-cause mortality Although an abnormal ECG in
al. 2014 (21) observational study, aimed Patients in the GESINUR patients with syncope is a common
24993462 at studying the association study who had syncope Results: Abnormal ECGs in 344 finding, only the presence of atrial
between specific ECG and had available, patients (65.6%). fibrillation, intraventricular
abnormalities and readable ECG and 12 mo 33 Patients died during follow-up conduction disturbances, left
mortality in patients with follow-up data (6.3%): ventricular hypertrophy ECG criteria,
syncope from the GESINUR - 1 due to SCD and ventricular pacing is associated
study. - Atrial fibrillation (OR: 6.8; 95% with 1-year all-cause mortality.
CI: 2.816.3, p<0.001)
Size: 524 patients - intraventricular conduction
disturbances (OR: 3.8; 95% CI:
1.78.3; p=0.001),
- LV hypertrophy ECG criteria
(OR: 6.3, 95% CI: 1.526.3;
p=0.011)
- ventricular pacing (OR 21.8,
95% CI 4.1115.3, P <.001)

13

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 3. RCTs Comparing Ambulatory Electrocardiography (Section 4.2.2)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if any);
Author; Study Type; (# patients) / (Absolute Event Study Limitations;
Year Published Study Size (N) Study Comparator Rates, Adverse Events
(# patients) P values; OR or RR; &
95% CI)
Barrett et al. Aim: Compare Inclusion criteria: patients Intervention: 24 h 1 endpoint: Prolonged duration
2014 (22) Holter to a 14 d for evaluation of cardiac Holter and 14 d Adhesive 96, Holter 61 monitoring for detection of
24384108 patch electrode arrhythmia adhesive patch events (p<0.001) arrhythmia events using single
lead, less-obtrusive,
Study type: Head Exclusion criteria: skin Comparator: Adhesive-patch monitoring
to head allergies, conditions, or Detection of platforms could replace
comparison, sensitivities to any of the arrhythmia over total conventional Holter monitoring
simultaneous components of wear time. in patients referred for
the adhesive patch Any 1 of 6 ambulatory ECG monitoring.
Size: monitor, receiving or arrhythmias, including
146 pt anticipated to receive supraventricular
pacing or external direct tachycardia,
current cardioversion, or AF/flutter, pause
the anticipation of being greater than 3s, AV
exposed to high-frequency block, VT, or
surgical equipment during polymorphic VT/VF.
the monitoring period
de Asmundis et Aim: head to Inclusion criteria: Intervention: 24 h 1 endpoint: Clinical Longer time and patient-
al. 2014 (23) head comparison Indication for monitor monitor and 15 d diagnosis for activated monitor improved
24574492 of 24 h Holter (palpitations 92.3%, HeartScan symptoms: yield. This was NOT a loop
and hand held dizziness 7.7%) Holter 1.8% recorder
patient-activated Comparator: Percent HeartScan 89%
even monitor Exclusion criteria: diagnosis of symptom- (p<0.01)
(not loop) presence of a pacemaker related arrhythmias
or an ICD, syncope,
Study type: structural heart
Sequential diseases, ECG
comparison abnormalities, and a Hx of
(Holter, then documented arrhythmia.
monitor)

14

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size:
625

Data Supplement 4. Nonrandomized Trials, Observational Studies, and/or Registries of Ambulatory Monitors (Section 4.2.2)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Turakhia et al. 2013 Study type: Inclusion criteria: Zio 1 endpoint: evaluated compliance, Demonstrates yield and compliance
(24) observational placed analyzable signal time, interval to with patch monitor although VT/VF
23672988 arrhythmia detection, and diagnostic not a major issue here
Size: 26,751 Exclusion criteria: yield of the Zio Patch,
N/A COMPARED: first 48h with later (mean
7.6 d)

Results:
Any arrhythmia: 62.2% vs 43.9%
Symptomatic arrhythmia: 9.7% vs 4.4%
VT 187 pt (0.7%)
PMVT, TdP, VF 6 pt (0.0%)
Linzer et al. 1990 Study type: Inclusion criteria: 1 endpoint: Monitor up to 1 mo with 25% yield for syncope Dx after
(25) observational Syncope with negative Loop negative Holter
2371954 Holter VT/VF uncommon (1 pt)
Size: 57 Results: arrhythmia was the cause of
Exclusion criteria: symptoms (diagnostic
Patients who had yield 25%; 95% CI: 1438%).
undergone EPS VT (1 patient), high grade AV block
(2 patients), SVT (1 patient),
asystole or junctional bradycardia from
neutrally mediated syncope (3
patients) and normal cardiac
rhythms (the remaining 7 patients).

15

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 5. Nonrandomized Trials, Observational Studies, and/or Registries of Implanted Cardiac Monitors (Section 4.2.3)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Turakhia et al. Am J Study type: Inclusion criteria: Zio placed 1 endpoint: evaluated Demonstrates yield and compliance
Car 2013 (24) observational compliance, analyzable with patch monitor although VT/VF
23672988 Exclusion criteria: N/A signal time, interval to not a major issue here
Size: 26,751 arrhythmia detection, and
diagnostic yield of the Zio
Patch
COMPARED: first 48 h with
later (mean 7.6 d)

Results:
Any arrhythmia: 62.2% vs
43.9%
Symptomatic arrhythmia:
9.7% vs 4.4%
VT 187 pt (0.7%)
PMVT, TdP, VF 6 patients
(0.0%)
CARISMA Study type: Inclusion criteria: AMI and 1 endpoint: incidence and Intermittent AV block was
Bloch Thomsen et al. observational reduced LVEF prognostic significance of associated with very high risk of
2010 (26) arrhythmias post MI with cardiac death
20837897 Size: Exclusion criteria: Refusal; reduced LVEF
297 participants inability of the patient to
participate in the study Results:
because of other serious Brady and
illness (N=312), tachyarrhythmias seen in
planned coronary bypass graft 137 patients (46%), with
surgery (N=184), or death 86% asymptomatic. 13%
(N=89). incidence of NSVT (16 bts),
3% sustained VT (30 sec),
3% VF (16 bts). Also 28%
AF with fast vent response;
10% high degree AV block;

16

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
7% sinus brady, 5% sinus
arrest
Linzer et al. 1990 Study type: Inclusion criteria: Syncope 1 endpoint: Monitor up to 25% yield for syncope diagnosis
(25) observational with negative Holter 1 mo with Loop after negative Holter
2371954
Size: Exclusion criteria: Prior EPS. Results: arrhythmia was
57 participants the cause of symptoms
(diagnostic yield 25%; 95%
CI 1438%). VT (1 patient),
high grade AV block (2
patients), SVT (1 patient),
asystole or junctional
bradycardia from neutrally
mediated syncope (3
patients) and normal
cardiac rhythms (the
remaining 7 patients).
Volosin et al. 2013 Study type: Inclusion criteria: Patients 1 endpoint: Evaluate Sensitivity is high (96.5% or 99.3% if
(27) Observational, for who transmitted data studied tachycardia detection of programmed for slower VT.
23439867 CareLink monitoring with induced VA at time of device and software Shows excellent detection in
service ICD implant testing. artificial environment.
Size: Results: 15.1% had VT or
2190 patients overall Exclusion criteria: Patients FVT detected, although true
who transmitted data. who did not transmit over 4 VT was seen in only 10.4%.
Also studied induced mo period For induced 1909
arrhythmias tachycardia episodes
reviewed. Sensitivity of
VT/VF was 99.3%
Krahn et al. 1999 Study type: Inclusion criteria: recurrent 1 endpoint: Detection of Demonstrates utility of loop
(28) Observational undiagnosed syncope arrhythmias related to although no VT/VF seen in this
9918528 recurrent syncope, with relatively small study.
Size: Exclusion criteria: unlikely to prior Holter
85 survive 1y, were unable to
give informed consent, had a Results: 68% had syncope.
previously implanted Arrhythmia seen in 42%
programmable medical who transmitted rhythm
device, were pregnant, or during symptoms.

17

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
were women of childbearing Bradyarrhythmia in 18,
potential not on a reliable tachyarrhythmia in 3 (SVT 2,
form of contraception. AFL 1; no VT/VF)

Solbiati et al. 2016 Study type: Inclusion criteria: 1 endpoint: To assess the This confirmed the advantage of the
(29) Systematic review, Unexplained Recurrent incidence of mortality, QoL, ILR in making a diagnosis in
27092427 Meta-analysis Syncope, evaluation of loop adverse events and costs of unexplained syncope, with trend seen
recorder vs no loop recorder ILRs vs. conventional in reduction of relapse.
Size: diagnostic workup in people
579 participants in 4 Exclusion criteria: N/A with unexplained syncope
trials
Results: No difference in
long-term mortality

2 studies showed trend of


reduction in syncope
relapse after diagnosis with
the ILR

Higher rate of diagnosis (RR:


0.61; 95% CI: 0.540.68)

Data Supplement 6. Nonrandomized Trials, Observational Studies, and/or Registries Comparing Noninvasive Cardiac Assessment (Section
4.2.4)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if
Author; Study Type; (# patients) / (Absolute Event Rates, any);
Year Published Study Size (N) Study Comparator P values; OR or RR; & Study Limitations;
(# patients) 95% CI) Adverse Events
VALIANT Aim: To evaluate Inclusion criteria: Intervention: 1 endpoint: The risk of Each 5% lower LVEF was
Solomon et risk and predictors of patients with first or Analysis of rates of sudden death was greatest in associated with a 21%
al. 2005 (30) SCD in patients post subsequent MI with HF, SCD. Evaluation of the first 30 d after MI: 1.4% increase in adjusted risk of
15972864 MI with left LV dysfunction, or both EF determined by per mo, 95% CI: 1.2%1.6% SCD or CA with resuscitation.
ventricular echocardiography and decreased to 0.14% per
dysfunction and/or Exclusion criteria: ICD as well as other mo 95% CI: 0.11%0.18%
HF in place prior to parameters. after 2 y after MI. Patients
randomization
18

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: Comparator: with LVEF <30% were at the
Observational study N/A greatest risk for SCD
of patients enrolled
in a RCT

Size: 14,609 patients


SCD-HEFT Aim: To determine Inclusion criteria: Intervention: Type 1 endpoint: Multivariable Among HF patients with an
Gula et al. with baseline Patients with HF, NYHA of modality to analysis showed that there LVEF between 20% and 35%,
2008 (31) assessment of EF class II-III and LVEF evaluate LVEF and was no significant difference each 5% increase in LVEF was
19033019 being performed 35% clinical outcomes. in survival between patients associated with a lower
using enrolled based on LVEF mortality risk (HR: 0.81; 95%
echocardiography, Exclusion criteria: Comparator: N/A determined RNA vs. CI: 0.750.88). The findings
RNA, or contrast Contraindication to echocardiography (HR: 1.06; were similar for each initial
angiography amiodarone or 1 95% CI: 0.881.28), RNA EF imaging modality, with the
impacted the prevention ICD Vs. angiography (HR: 1.25; interaction term combining
likelihood of survival. 95% CI: 0.971.62), or imaging method and LVEF in
echocardiography vs. the Cox model was NS
Study type: angiography (HR: 1.18; 95% CI: (p=0.71).
Observational 0.941.48).
analysis of patients
enrolled into a RCT

Size: 2,521 patients

Data Supplement 7. Nonrandomized Trials, Observational Studies, and/or Registries Comparing Biomarkers (Section 4.2.5)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if any);
Author; Study Type; (# patients) / (Absolute Event Rates, Study Limitations;
Year Published Study Size (N) Study Comparator P values; OR or RR; & Adverse Events
(# patients) 95% CI)
Korngold et al. Aim: Evaluate Inclusion criteria: Intervention: NT- 1 endpoint: Relationship Women with NT-proBNP
2009 (32) baseline NT-proBNP Women nurses 3055 y proBNP data at of NT-proBNP and SCD levels above the cut point of
19470888 levels to predict risk of age baseline. 99 SCD (RR for 1-standard 389 pg/mL were at increased
of SCD in a general cases and 294 deviation increment 1.49; risk of SCD (RR 5.68; 95% CI:
population of Exclusion criteria: matched controls. 95% Cl: 1.092.05; 1.7818.2; p=0.003).
women. Blood sample not p=0.01)
collected Comparator: N/A

19

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: Case
Control

Size: 32,826 women


with biomarker data
out of 121,700
enrolled
Patton et al. Aim: Evaluate risk of Inclusion criteria: Men Intervention: NT- 1 endpoint: Higher NT- NT-proBNP was associated
2011 (33) SCD as function of and women 65 y of age proBNP levels were proBNP levels were with SCD after adjustment for
21044699 baseline NT-proBNP and older randomly analyzed both as a associated with SCD, with clinical characteristics and risk
in a community selected from 4 continuous an unadjusted HR: 4.2; factors (age, sex, race, and
cohort of older men communities variable, where the 95% CI: 2.9, 6.1; p=0.001 other associated conditions),
and women natural log of NT- for the highest vs. lowest with an adjusted HR for the
Exclusion criteria: NT- proBNP was used, quintile fifth vs. the first quintile of 2.5
Study type: Cohort proBNP levels not and as categorized (95% CI: 1.6, 3.8; p=0.001).
study available into quintiles

Size: 5,447 men and Comparator: N/A


women
Scott et al. Aim: Evaluate Inclusion criteria: Intervention: BNP 1 endpoint: Increased The risk of SCD associated
2009 (34) whether BNP levels Studies evaluating BNP and NT-proBNP levels BNP or NT-proBNP with increased BNP or NT-
19789399 can predict SCD and or NT-proBNP levels for evaluated for SCD risk predicted SCD with a RR: proBNP tended to be higher in
VA in patients SCD or VA in patients without 3.68; 95% CI: 1.907.14 in patients with a lower LVEF.
without ICDs ICD or VA risk in patients without ICDs. However, there was not a
Exclusion criteria: patients with ICD Increased BNP or NT- significant interaction between
Study type: Meta- Overlapping studies proBNP predicted VA with BNP level and LVEF on risk
Analysis of Comparator: N/A a RR: 2.54; 95% CI: 1.87 prediction.
Observational 3.44.
Studies

Size: 14 studies (6
studies with 3,543
patients without ICD
and 8 studies of
1,047 patients with
ICD)

20

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Blangy et al. Aim: Evaluate Inclusion criteria: Intervention: Serum 1 endpoint: In a In addition, LVEF <0.35 (OR
2007 (35) biomarkers on VT Patients with BNP, hs-CRP, and multivariate analysis, an 2.19; 95% CI: 1.004.79) was
17526509 risk in patients with spontaneous sustained procollagen levels increased serum BNP (OR: associated with a higher VT
ICD post MI VT post MI receiving measures at baseline 3.75; 95% CI: 1.469.67), incidence.
ICD an increased hs-CRP (OR:
Study type: Comparator: N/A 3.2; 95% CI: 1.268.10,
Observational Exclusion criteria: N/A and an increased PINP
(OR: 3.71; 95% CI: 1.40
Size: 121 men and 9.88 were associated with
women a higher VT incidence.

HF ACTION Aim: Evaluate Inclusion criteria: Intervention: NT- 1 endpoint: Elevations NT-proBNP was more
Ahmad et al. biomarkers in NYHA class II to IV proBNP, galectin-3, in each biomarker was strongly predictive of pump
2014 (36) prediction of sudden chronic HF with and ST2 levels were associated with increased failure (C statistic: 0.87)
24952693 deathand EF35% assessed at baseline risk for SCD death in both Addition of ST2 and
progressive HF death in patients enrolled in adjusted and unadjusted galectin-3 led to improved net
in patients with HF Exclusion criteria: the trial of exercise analyses. risk classification of 11% for
with reduced EF Biomarker data not training vs. usual care However, increases in the SCD.
obtained biomarkers had stronger There was no improvement
Study type: Inability to exercise Comparator: N/A associations with pump in net risk reclassification for
Observational failure than SCD. Clinical pump failure death with ST2 or
analysis of subjects variables along with NT- galectin-3
enrolled in a RCT proBNP levels were
predictors sudden cardiac
Size: 813 subjects death (C statistic: 0.73).
Levine et al. Aim: To evaluate Inclusion criteria: BNP Intervention: BNP or 1 endpoint: In Quartile analyses showed
2014 (37) the ability of BNP or or NT-proBNP levels NT-proBNP levels to multivariate analysis NT- higher relative risk of VA
24837348 NT-proBNP to and 1 prevention ICD predict risk of VA proBNP was associated compared to the relative risk
predict VA in with increased risk of VA of all-cause mortality for both
patients with 1 Exclusion criteria: BNP Comparator: N/A with HR: 5.75; p<0.001 BNP and NT-proBNP.
prevention ICDs or NT-proBNP not and BNP was associated
available within 12mo with increased risk with
Study type: of ICD implantation. HR: 3.40; p<0.01.
Observational

Size: 564 patients

21

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Berger et al. Aim: To evaluate Inclusion criteria: Intervention: BNP 1 endpoint: In Using a cutoff point of log
2002 (38) role of BNP in Patients with HF and levels at baseline and multivariate analysis, log BNP 2.11 (130 pg/mL), the KM
12021226 predicting SCD in reduced EF with BNP association with BNP level was the only sudden deathfree survival
patients with HF level measured at subsequent SCD independent predictor of rates were significantly higher
with LVEF 35% baseline sudden death in patients below (99%)
Comparator: N/A compared with patients above
Study type: Exclusion criteria: (81%) this cutoff point
Observational Patients with heart (p=0.0001)
transplantation or VAD
Size: 452 patients

Data Supplement 8. RCTs Evaluating EP Study for VA (Section 4.3.2)


Study Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if any);
Acronym; Study Type; (# patients) / (Absolute Event Rates, Study Limitations;
Author; Study Size (N) Study Comparator P values; OR or RR; & Adverse Events
Year Published (# patients) 95% CI)
Buxton AE, et Aim: to analyze the Inclusion criteria: CAD, Intervention: AAD 1 endpoint: 61% of events were
al. Circ 2002 relationship of EF EF<40%, and or ICD for Total mortality and arrhythmic among inducible
(39) and inducible VA to asymptomatic, inducible patients arrhythmic deaths/cardiac patients with EF 30% and only
12417544 mode of death unsustained VT arrests more common in 42% among noninducible
Comparator: EF patients with EF <30% patients, p=0.002
Study type: Exclusion criteria: History 3040% vs. <30% Arrhythmic deaths
Prospective, of syncope, sustained similar in patients with EF
randomized, RCT VT/VF more <30% and 3040%
than 48 h after AMI, Relative contribution of
Size: 1791 patients unsustained VT arrhythmic deaths to total
only in the setting of drug- mortality was higher in
induced LQTS or AMI or inducible patients (58% of
that was attributable deaths vs. 46% of deaths
to acute metabolic in noninducible patients,
disorders or drug toxicity, p=0.004
or symptomatic,
unsustained VT

22

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
MUSTT Aim: to test the Inclusion criteria: CAD, ntervention: AAD 1 endpoint: CA or The risk of cardiac arrest or
Buxton AE, et usefulness of EPS for EF<40%, and or ICD arrhythmic death death from arrhythmia among
al NEJM 1999 risk stratification for asymptomatic, At 5 y, inducible patients who received
(40) SCD unsustained VT Comparator: patients treated with treatment with ICDs was
10601507 Patients with AAD/ICD had a significantly lower than that
Study type: Exclusion criteria: History inducible VT/VF at significantly lower risk of among the patients discharged
Prospective, of syncope, sustained EPS randomized to arrhythmic death or CA without receiving defibrillator
randomized, RCT VT/VF more treatment with (25%) than patients not treatment (RR: 0.24; 95% CI:
than 48 h after AMI, AAD or ICD vs. no receiving antiarrhythmic 0.130.45; p<0.001).
Size: 704 patients unsustained VT specific therapy (32%) (RR: 0.73; Reduction in 1 endpoint in
with inducible, only in the setting of drug- antiarrhythmic 95% CI: 0.530.99) AAD/ICD arm was due to
sustained VA induced LQTS or AMI or treatment reduction in events in patients
that was attributable treated with ICDs, not AAD
to acute metabolic
disorders or drug toxicity,
or symptomatic,
unsustained VT
MUSTT Aim: to test the Inclusion criteria: CAD, EF Intervention: EPS 1 endpoint: CA or Patients with ischemic
Buxton et al. usefulness of EPS for <40%, and asymptomatic, arrhythmic death cardiomyopathy and
2000 (41) risk stratification for unsustained VT Comparator: At 2 and 5 y, noninducible asymptomatic, unsustained VT
10874061 SCD Inducible VT/VF at patients had a with inducible VT have a
Exclusion criteria: History EPS and not significantly lower risk of significantly greater risk of SCD
Study type: of syncope, sustained treated with AAD arrhythmic death or CA or CA and higher overall
Prospective, VT/VF more or ICD compared (12%, 24%) than inducible mortality than similar patients
randomized, RCT than 48 h after AMI, to noninducible patients (18%. 32%) who are noninducible
unsustained VT patients (adjusted p<0.001).
Size: 1750 patients only in the setting of drug- Overall mortality at 5 y
(353 inducible; 1397 induced LQTS or AMI or was lower in noninducible
noninducible) that was attributable patients (44% vs. 48%,
to acute metabolic adjusted p=0.005).
disorders or drug toxicity,
or symptomatic, Safety endpoint (if
unsustained VT relevant): N/A

23

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
MADIT-I Aim: To evaluate Inclusion: Previous MI, Comparator: All-cause mortality: In patients with a prior MI,
Moss et al. whether LVEF 35%, NSVT, Control (101 Control 32% vs. ICD 13% low EF who are at high risk for
1996 (42) prophylactic ICD, as inducible VT at EPS that patients) (RRR -59% ARR -19%) VT, prophylactic therapy with
8960472 compared with was non-suppressed with an ICD leads to improved
conventional IV procainamide or Intervention: survival as compared with
medical therapy, equivalent AAD ICD (95 patients) conventional medical therapy.
would improve
survival in a high-risk Exclusion: previous CA or
group of patients VT causing syncope that
with NSVT, reduced was not associated with an
LVEF and previous AMI; symptomatic
MI. hypotension while in a
stable rhythm; and MI <3
Study type: wk, prior CABG <2 mo or
prospective PCI <3 mo, as were
multicenter RCT women of childbearing
age who were not using
Size: 196 patients medically prescribed
contraceptives, patients
with advanced
cerebrovascular disease,
patients with any
condition other than
cardiac disease that was
associated with a reduced
likelihood of survival for
the duration of the trial,
and patients who were
participating in other
clinical trials
SCD-HeFT Aim: Evaluate Inclusion: NYHA class I-III Intervention 1: All-cause mortality: In patients with NYHA class II
Bardy et al. whether HF, LVEF35% GDMT plus a ICD control 36% vs. ICD 29% or III HF and LVEF35%,
2005 (43) amiodarone or a (829 patients) (RRR -23% and ARR -7%) amiodarone has no favorable
15659722 conservatively Exclusion: <18 y, unable to effect on survival, whereas
programmed shock- give consent Intervention 2: single-lead, shock-only ICD
only, single-lead ICD therapy reduces overall
would decrease the

24

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
risk of death from GDMT plus mortality. This was the longest
any cause in a broad amiodarone (845 and largest ICD trial.
population of patients)
patients with mild-
to-moderate HF Comparator 1:
GDMT plus
Study type: Placebo (847
prospective patients)
multicenter RCT

Size: 2521 patients


MADIT-II Aim: To evaluate Inclusion: Prior MI (>1 Comparator: All-cause mortality: In patients with a prior MI
Moss et al. the benefit of ICD in mo), EF 30% Control (490 control 22% vs. ICD 16% and advanced left ventricular
2002 (44) patients with prior patients) (RRR -28% and ARR -6%) dysfunction, prophylactic ICD
11907286 MI and reduced Exclusion: existing improves survival and should be
LVEF indication for ICD; NYHA Intervention: considered as a recommended
class IV at enrollment; had ICD (742 patients) therapy.
Study type: RCT undergone coronary
revascularization <3 mo;
Size: 1232 patients MI <30 d, advanced
cerebrovascular disease,
childbearing age and not
using contraceptive,
presence of any condition
other than cardiac disease
that was associated with a
high likelihood of death
during the trial, or
unwilling to provide
consent

25

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 9. Nonrandomized Trials, Observational Studies, and/or Registries of EP Study for VA - (Section 4.3.2)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Hilfiker et al. 2015 Study type: Inclusion criteria: Patients 1 endpoint: SCD or appropriate Mixed population of patients
(45) prospective cohort who underwent EPS for SCD ICD therapy EPS identifies patients who are likely
26131339 risk evaluation because of to have recurrent VA or SCD.
Size: 265 patients structural or functional Results: Sustained VT was
heart disease and/or induced in 125 patients (47.2%)
electrical conduction and non-sustained VT in 60
abnormality and/or after patients (22.6%)
syncope/CA. 153 patients (57.7%) underwent
ICD implantation
Exclusion criteria: Not 1 endpoint event occurred in 49
specified patients (18.5%).
Cox regression analysis showed
that both sustained VT during
EPS (HR: 2.26; 95% CI: 1.224.19,
p=0.009) and EF<5% (HR: 2.00;
95% CI: 1.133.54, p=0.018) were
independent predictors of 1
endpoint events.
Bourke et al. 1991 Study type: Inclusion criteria: recent 1 endpoint: documented EPS predicts VT/VF in follow-up of
(46) prospective cohort AMI sustained VT/VF or witnessed survivors of AMI
1907984 sudden death
Size: 1209 patients Exclusion criteria: early
recurrence of angina Results: Sustained monomorphic
requiring treatment; VT was inducible by programmed
spontaneous VT or VF more electrical stimulation in 75
than 48 h after MI; CHF not (6.2%).
controlled with furosemide; 14 infarct survivors (19%) with
significant noncardiac inducible VT experienced
disease spontaneous VT or VF compared
with 34 (2.9%) of those without
inducible VT (p<0.0005).

26

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Bailey et al. 2001 Study type: meta- Inclusion criteria: 44 reports 1 endpoint: sustained VT/VF, Multiple tests evaluated: SAECG;
(47) analysis for which incidence of major CA, sudden death heart rate variability; severe VA on
11738292 arrhythmic events and ambulatory electrocardiography; EF;
Size: 4022 post-MI predictive accuracy could be Results: positive EPS had 61.6% and EPS.
patients inferred sensitivity and 84.1% specificity Results for all tests evaluated were
2 y probability of event was similar
Exclusion criteria: N/A 25.5% EPS has moderate predictive value for
RR 6.6; OR 8.5 life-threatening VA.

Schmitt et al. 2001 Study type: Inclusion criteria: post-MI 1 endpoint: sudden death, A subgroup of 96 high-risk patients
(48) prospective cohort patents identified as high symptomatic VT, CA declined
11401129 risk by scoring system EPS. In this non-consent group,
Size: 98 post-MI including EF, PVCs, and Results: Patients underwent EPS. cardiac mortality (combined sudden
patients identified abnormal SAECG Event rate was 33% with a and nonsudden) was significantly
as high risk by positive EPS vs. 2.6% (p<0.0001) higher (log-rank chi-square 9.38 RR 4.7;
noninvasive Exclusion criteria: Hx of with a negative EPS. 95% CI: 1.613.9, p=0.0022) compared
markers spontaneous sustained VT to group treated according to results of
EPS.
20/21 patients with a positive EPS had
ICD implanted.

Brembilla-Perrot et Study type: Inclusion criteria: EF<40% 1 endpoint: cardiac mortality EPS may be useful to determine
al. 2004 (49) Prospective and syncope mechanism of syncope in patients with
15358027 observational Results: Sustained VT was ischemic cardiomyopathy.
Exclusion criteria: unstable induced in 44 group I patients
Size: 180 patients angina; recent AMI; recent (37%) and 13 group II patients
(119 CAD, group 1; coronary angioplasty or (21%); VFL (>270 beats/min) or
61 DCM, group 2) CABG; second- or third- VF was induced in 24 group I
degree AV block; sustained patients (19%) and 9 group II
supraventricular or patients (15%)
ventricular arrhythmia; VT or VF induction was predictive
clinical HF not controlled by of mortality in CAD and identified
furosemide; uncontrolled a group with high cardiac
electrolyte abnormalities; mortality (46%), compared with
significant noncardiac patients with a negative study,
disease; or amiodarone who had a lower mortality (6%;
treatment.

27

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
p<0.001). Cardiac mortality was
only correlated with EF in DCM.
Bhandari AK Circ Study type: Inclusion criteria: LQTS 1 endpoint: EP testing in LQTS Inducibility of nonsust VT did not
1985 (50) retrospective single with syncope or ACA provide prognostic information.
2856866 center Mean QTc 550 msec Results: RV and LV EPS, 3 EP studies of limited value in
extrastimuli, with and without diagnosis, treatment of LQTS patients.
Size: 15 11 control subjects, normal isuprel
QTc Rapid polymorphic VT: 40%
Exclusion criteria: No pt with inducible sustained VT
N/A or VF
Giustetto C EHJ Study type: Inclusion criteria: Short QTc 1 endpoint: outcomes with Short QTS may be a cause of SCD in
2006 (51) Retrospective single 340 msec and personal or AICD or hydroquinidine infancy
16926178 center family Hx of CA. 73% males. Hydroquinidine may be proposed in
Results: Median age dx 30y (4- children or patients not suitable for
Size: 29 Exclusion criteria: N/A 80); 62% symptomatic: syncope AICD
24%, AF 31%. 34% ACA (10 PES sensitivity 50%
patients); 2/10 had CA in infancy.
In 28% ACA was initial symptom.
ICD implanted in 14; 10
hydroquinidine. Median followup
23 mo (9-49), one pt with
appropriate ICD shock. No pt on
hydroquinidine had SCD or
syncope.

ES 18/29: Ventricular ERP 140-


180 msec. VF induced in 61%
(11/18); 3/6 with documented VF
had inducible VF: sensitivity 50%.
AERP CL 600: 120-180 ms, mean
157.
Mahida S JACC Study type: Inclusion criteria: Patients 1 endpoint: Inducibility of VF in EPS not useful to risk stratify patients
2015 (52) multicenter with ER and ACA due to VF patients with ACA and ER on ECG with prior VF arrest and ER
25593056 observational underwent EPS. Mean age and outcomes. Followup 74.9 y
36 13y. Followup with ICD
Size: 81 interrogations. Results: VF inducible in 22%.

28

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: N/A Recurrent VF in 33% of inducible
VF, vs. 33% of those with non-
inducible VF. p=NS, 0.93.
VF inducibility did not correlate
with max J wave amplitude or
distribution

Giustetto C JACC Study type: Inclusion criteria: European 1 endpoint: syncope, CA or SQTS assoc with SCD in all ages
2011 (53) retrospective multi- Short QT Registry patients approp ICD shocks SQTS Symptomatic patients have high risk
21798421 center with QTc 360 msec with Hx of recurrent arrhythmic events
sudden death, ACA, Results: Mean Followup 6427 Patients treated with Hydroquinidine
Size: 53 syncope; mo. Median age 26 y (IQR 17 did not have arrhythmic events
patients with QTc 340 39). 62% symptomatic: 32% with Asymptomatic patients: no CA/ICD
msec included without ACA (13 patients) or sudden shocks.
symptoms. death (4), syncope (8), AF (6), PES not sensitive
palpatations (13).
75% males. Age at CA 3 mo62 y.
Males: >90% of CA occurred
Family Hx SCD/CA (11). between 1440 y.
Genotype positive 23% of Prevalence CA males 35%,
probands: HERG in 4 females 30%.
families (N588K in 2, T6181 AICD in 24, hydroquinidine in 12.
in 2; CACNB2b in one family) 11/12 with prior CA received ICD:
2 approp ICD shocks. 58%
Exclusion criteria: N/A complications of ICD, inapprop
shocks due to T wave
oversensing 4/14.
PES: 28 patients. VERP CL 600-
500: mean 166 msec. AERP 166
msec. VF induced in 16/28: 3/28
with prior CA = sensitivity 37%,
NPVs 58%.
Overall event rate 3.3%/y: 4.9%
in patients without AA drugs.
Asymptomatic patients: 27. ICD
implanted in 9 due to + family Hx
or induced VF. Two long term

29

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
quinidine. One syncope; 2
nonsust VT on ICD.
Raczak et al. 2004 Study type: Inclusion criteria: post-MI 1 endpoint: appropriate ICD 97 patients had ICDs implanted
(54) prospective cohort patients with documented shock or sudden or unwitnessed EPS not useful in predicting
15226627 VF, sustained VT, or syncope death arrhythmias in follow-up
Size: 112 patients and NSVT
Results: Sustained VT induced in
Exclusion criteria: AF, SND 84% and 77% of patients who did
or AV block, insulin- or did not develop arrhythmia in
dependent DM, frequent follow-up (p=0.34)
(>5%) ectopic beats Baroreflex sensitivity <3.3
ms/mmHg was only predictor of
arrhythmia recurrence in
patients with EF <35% (sensitivity
79%, specificity 74%, positive and
NPVs 83% and 68%)

AVID Study type: Inclusion criteria: patients 1 endpoint: death or recurrent EPS is of limited value in patients with
Brodsky et al. 2002 substudy from with VF, VT with syncope, or VT/VF a Hx of sustained VA.
(55) prospective clinical sustained VT in the setting
12228785 trial of LV dysfunction who Results: 384 (67%) had inducible
underwent EPS sustained VT or VF. Inducible
Size: 572 patients patients were more likely to have
Exclusion criteria: N/A CAD, previous infarction, and VT
as their index arrhythmic event.
Inducibility of VT or VF did not
predict death or recurrent VT or
VF.

MADIT II Study type: Inclusion criteria: Patients 1 endpoint: sustained VT/VF ICD therapy for spontaneous VF was
Daubert et al. 2006 substudy from from MADIT II (previous MI, less common (p=0.021) in inducible
(56) prospective clinical EF<30%) who received ICDs Results: The 2 y KM event rate patients than in noninducible patients.
16386671 trial and underwent EPS for VT or VF was 29.4% for
inducible patients and 25.5% for
Size: 593 patients Exclusion criteria: control noninducible patients (p=0.280,
patients; ICD patients with by log-rank analysis).
no EPS

30

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Inducible patients had a greater
likelihood of experiencing ICD
therapy for VT than noninducible
patients (p=0.023).

ABCD Study type: Inclusion criteria: ischemic 1 endpoint: appropriate ICD Both tests somewhat helpful in risk
Costantini et al. Prospective cohort; cardiomyopathy (EF <40%) discharge or sudden death stratification, but NPV is not 100%
2009 (9) patients underwent and NSVT
19195603 EPS and T wave Results: 39 (7.5%) met the 1
alternans testing; Exclusion criteria: unstable end point at 1y
ICDs were CAD; NYHA class IV; prior T wave alternans achieved 1 y
implanted if either CA, sustained VT, or positive (9%) and negative (95%)
test was positive unexplained syncope; <28 d predictive values comparable to
from MI, CABG, or PCI; EPS (11% and 95%).
Size: 566 patients permanent AF; on an AAD. Event rate with both tests
negative was 2% vs. 12% with
both tests positive (p=0.017).
DEFINITE Study type: Inclusion criteria: dilated 1 endpoint: appropriate ICD Inducibility of either VT or VF was
Daubert et al. 2009 substudy of cardiomyopathy (EF<35%), therapy for VT/VF or arrhythmic associated with an increased likelihood
(57) DEFINITE NSVT or frequent PVCs, and death of subsequent ICD therapy for VT or VF.
19545338 NYHA class I-III, randomized
Size: 204 patients to ICD arm; noninvasive EPS Results: Inducibility was found in
performed through ICD 29 of 204 patients (VT in 13, VF in
16). 34.5% of the inducible group
Exclusion criteria: NYHA (10 of 29) experienced ICD
class IV or permanent therapy for VT or VF or
pacemaker arrhythmic death vs. 12.0% (21
of 175) of the noninducible
patients (HR: 2.60; p=0.014).

Gold et al. 2000 Study type: Inclusion criteria: patients 1 endpoint: SCD, sustained Both T-wave alternans testing and
(58) prospective, undergoing diagnostic EPS VT/VF or appropriate ICD therapy EPS predicted VT.
11127468 multicenter who were in sinus rhythm
and able to do bicycle Results: KM survival analysis of
Size: 215 patients exercise; reasons for EPS the 1 end point showed that T-
included syncope, CA, wave alternans predicted events
sustained VT, SVT

31

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
with a RR:10.9; EPS had a RR: 7.1;
Exclusion criteria: not and SAECG had a RR: 4.5.
specified Multivariate analysis of 11
clinical parameters identified
only T-wave alternans and EPS as
independent predictors of
events.

Gatzoulis et al. Study type: Inclusion criteria: 1 endpoint: total mortality and EPS inducibility of sustained VT/VF is
2013 (59) prospective cohort symptomatic idiopathic appropriate ICD activation predictive of future ICD activation but
23588627 DCM >6 mo not total mortality in patients with CDM
Size: 158 patients Results: EPS performed in all
Exclusion criteria: Hx of patients; 44 (27.8%) had
aborted SCD or sustained inducible VT/VF.
VT; NYHA class IV; Hx of MI ICDs implanted in 41/44
or myocarditis; significant inducible patients and 28/114
VHD; hypertrophic or noninducible patients.
restrictive cardiomyopathy; No difference in mortality
alcohol-associated disease; Inducibility was associated with
cardiac toxicity ICD activation events (30/41
inducible patients (73.2%) vs.
5/28 noninducible patients
(17.9%), p=0.001.

Data Supplement 10. RCTs for Preventing SCD with HF Medications - (Section 5.2)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint
Author; Study Type; (# patients) / (Absolute Event Rates, (if any);
Year Published Study Size (N) Study Comparator P values; OR or RR; & Study Limitations;
(# patients) 95% CI) Adverse Events
CAPRICORN Study type: RCT Inclusion criteria: Intervention: Carvedilol 1 endpoint: All-cause BB improve mortality
Dargie et al. 2001 Recent MI (3-12 d); EF up to 25mg BID mortality 12% vs 15%, HR: post-MI in patients
(60) Aim: to test <40% 0.77; 95% CI 0.600.98, with LV dysfunction
11356434 whether carvedilol Comparator: Placebo p=003).
added to standard Exclusion criteria VT/VF significantly
AMI care in reduced.

32

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
patients with left Uncontrolled HF, VT/VF: 3.9% vs. 0.9%. HR:
ventricular unstable angina, 0.24; 95% CI 0.110.49;
dysfunction would hypotension, p<0.0001.
improve outcomes. bradycardia

Size: 1959

US CARVEDILOL Study type: RCT Inclusion criteria: Intervention: Carvedilol 1 endpoint: survival and BB have a large
Packer et al. 1996 CHF, LVEF<35% Comparator: Placebo hospitalization effect on all cause and
(61) Aim: To determine - Mortality: 7.8% vs. 3.2 % SCD mortality.
8614419 the effects of Exclusion criteria - SCD 3.8% vs. 1.7%
carvedilol on Major procedure or
survival and surgery within 3 mo.
hospitalization

Size: 1094

CIBIS II Study type: RCT Inclusion criteria: Intervention: Bisoprolol 1 endpoint: all-cause Bisoprolol reduces
No Authors listed EF <35%, class III, IV, mortality all-cause mortality and
(62) Aim: To investigate standard therapy, Comparator: Placebo mortality from SCD.
10023943 the efficacy of CIBIS-II was stopped early,
bisoprolol in Exclusion criteria All-cause mortality 11.8% vs
decreasing all- N/A 17.3%. p<0.0001.
cause mortality in
chronic HF SCD 3.6% vs 6.3% p=0.0011.

Size: 2647

MERIT HF Study type: RCT Inclusion criteria: Intervention: 1 endpoint: mortality and BB reduce mortality
Hjalmarson et al. NYHA class II to IV, Metoprolol succinate hospitalization (time to in patients with HF.
(63)2000 Aim: To examine EF<40%; optimum Comparator: Placebo event).
10714728 the effects of standard therapy.
metoprolol CR/XL - All-cause mortality: 34%
on mortality, Exclusion criteria - SCD: 41% RR

33

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
hospitalization, N/A
symptoms, and QoL
in patients with HF.

Size: 3991
V-HEFT-II Study type: RCT Inclusion criteria: Intervention: Enalapril 1 endpoint: mortality Enalapril in patients
Cohn et al. 1991 NYHA Class II-III Comparator: Isosorbide with HF reduces
(64) Aim: To better Dinitrite Mortality 18% vs. 25% mortality and SCD
2057035 define vasodilator p=0.016. compared to
therapy in HF Isosorbide Dinitrite
Exclusion criteria SCD: 14% vs. 23%, p<0.05
Size: 804 N/A favoring enalapril

Val-HeFT Study type: RCT. Inclusion criteria: Intervention: Valsartan 1 endpoint: all-cause ARB added to ACE-I
Cohn et al. 2001 NYHA II, III (added to ACE-I) mortality are not additionally
(65) Aim: To explore the Comparator: Placebo helpful
11759645 efficacy of the Exclusion criteria Result: no difference in all-
addition of ARB to N/A cause mortality.
ACE-I therapy.

Size: 5010

VALIANT Study type: RCT Inclusion criteria: Intervention: Valsartan 1 endpoint: all-cause or CV ARB added to ACE-I
Pfeffer et al. 2003 Post-MI, LVEF<35%. 160 BID mortality are not additionally
(66) Aim: To explore the Class I or II HF. Comparator: Valsartan helpful
14610160 effects of ARB 80 BD No difference in either all-
added to ACE-I Exclusion criteria cause or CV related
therapy. N/A Both added to enalapril mortality

Size: 14,703

ELITE Study type: RCT Inclusion criteria: Intervention: Losartan 1 endpoint: tolerability ARB better than ACE,
Comparator: Captopril measure

34

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Pitt et al. Lancet Aim: To determine NYHA II IV, EF <40%, Only ARB trial to
1997 (67) the relative efficacy age >65 y 2 measure: mortality show a difference in
9074572 of ACE vs. ARB in Exclusion criteria SCD.
HF N/A All-cause mortality 4.8% vs. Small trial,
8.7% (p=0.035) Mortality was a 2
Size: 722 end-point.
36% relative risk reduction
in SCD
ELITE II Study type: RCT Inclusion criteria: Intervention: Losartan 1 endpoint: all-cause There were no
Pitt et al. 2000 (68) Age >60 y, class II-IV Comparator: Captopril mortality and SCD significant differences
10821361 Aim: To confirm HF, EF <40%. in all-cause mortality or
whether losartan is all-cause mortality (11.7 vs sudden death or
superior to Exclusion criteria 10.4%) p=0.16 resuscitated arrests
captopril N/A or sudden death or
resuscitated arrests (9.0 vs
Size: 3152 7.3%) p=0.08
RALES Study type: RCT Inclusion criteria: Intervention: 1 endpoint: all-cause Spironolactone
Pitt et al. 1999 (69) Class III, IV HF, EF spironolactone mortality reduced all-cause
10471456 Aim: To explore <35%, mortality and SCD in
whether a Comparator: placebo Death: 46% vs. 35%. patients with HF.
mineralocorticoid Exclusion criteria p<0.001
antagonist could N/A SCD: 13% vs. 10%, p=0.02
reduce mortality in
patients with HF.

Size: 1663
EPHESUS Study type: RCT Inclusion criteria: Intervention: 1 endpoint: All-cause Eplerenone reduced
Pitt et al. 2003 (70) 3-14 d post-MI Eplerenone mortality. all-cause and SCD in
12668699 Aim: To determine LVEF <40% Comparator: Placebo patients with HF
the effect of Death: 14% vs. 17%. RR
eplerenone on Exclusion criteria 0.85, p=0.008.
mortality among Creatinine >2.5
patients with AMI SCD: 5% vs. 6% (p=0.03)
and LV dysfunction

Size: 6632 Safety endpoint (if


relevant):

35

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Hyperkalemia: 5.5%
eplerenone vs. 3.9%
Hypokalemia: 8.4%
eplerenone vs. 13.1%

EMPHASIS Study type: RCT Inclusion criteria: Intervention: 1 endpoint: composite Significant reduction
Zannad et al. 2011 Class II HF Eplerenone death and HF hospitalization on composite
(71) Aim: To evaluate EF <35% endpoint. Non-
21073363 the effect of Comparator: Placebo 1 composite endpoint: significant reduction in
eplerenone on Exclusion criteria 18.3% vs. 25.9% (p<0.001) SCD.
patients with AMI, NYHA III, IV, GFR
chronic systolic HF. <30 SCD: 4.4% vs. 5.5%, p=0.12

Size: 2737 Safety endpoint (if


relevant):
Hyperkalemia: 11.8% vs.
7.2%
PARADIGM Study type: RCT Inclusion criteria: Intervention: 1 endpoint: CV death (2
Desai et al. 2015 Class II-IV HF Eplerenone analysis exploring mode of
(72) Aim: 2 analysis of EF <40% death)
26022006 the original Guideline rec. med Comparator: Placebo
PARADIGM-HF trial therapy CV death: HR: 0.80; 95% CI
to explore mode of 0.720.89, p<0.001.
death. Exclusion criteria
AMI, NYHA III, IV, GFR Among CV deaths,
Size: 8399 <30 SCD: HR: 0.80; p=0.008

death due to worsening HF:


HR: 0.79; p=0.034

36

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 11. RCTs and Nonrandomized Trials, Observational Studies, and/or Registries Related to Surgery and Revascularization
Procedures (Section 5.5)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if any);
Author; Study Type; (# patients) / (Absolute Event Rates, Study Limitations;
Year Published Study Size (N) Study Comparator P values; OR or RR; & Adverse Events
(# patients) 95% CI)
STICH Aim: Cause of death Inclusion criteria: age Intervention: CABG CABG therapy tended to
Carson et al. analysis for the 462 18 y, CAD amenable to (plus medical reduce cardiovascular
2013 (73) deaths during the CABG, and LVEF 35% therapy) deaths (HR: 0.83; 95% CI:
24621972 original follow-up 0.681.03; p=0.09) and
period of a median Exclusion criteria: Comparator: medical significantly reduced the
of 56 mo of the left main coronary therapy alone most common modes of
parent trial that stenosis 50% or death: sudden death (HR:
compared CABG Canadian 0.73; 95% CI: 0.540.99;
plus medical Cardiovascular Society p=0.041) and fatal pump
therapy to medical III-IV angina while failure events (HR: 0.64;
therapy alone to receiving medical 95% CI: 0.411.00;
reduce death from therapy p=0.05). Time-dependent
any cause estimates indicated that
the protective effect of
Study type: RCT CABG principally occurred
Size: 1212 patients after 24 mo in both
categories.
STICHES Aim: Compare CABG Inclusion criteria: age Intervention: CABG 1 endpoint: lower Cardiac arrest outcomes:
Velazquez et plus medical 18 y, CAD amenable to (plus medical mortality with CABG Sudden/arrhythmic death
al. 2016 (74) therapy to medical CABG, and LVEF 35% therapy) (58.9%) than the medical 116 (19%) CABG, 154 (26%)
27040723 therapy alone to therapy (66.1%) group. medical therapy
reduce death from Exclusion criteria: Comparator: medical CABG vs. medical Within 30 d after
any cause left main coronary therapy alone therapy, HR: 0.84; 95% CI: randomization
stenosis 50% or 0.730.97; p=0.02 by log- CA requiring CPR, 25 (4%)
Study type: RCT Canadian rank test. CABG and 2 (0.3%) medical
Cardiovascular Society therapy.
Size: 1212 patients, III-IV angina while
with 9.8 y median receiving medical
followup therapy

37

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
AVID Registry Aim: determine Inclusion criteria: Intervention: Patients who underwent
Cook et al. whether patients Ventricular fibrillation revascularization; ICD revascularization
2002 (75) with CAD who or symptomatic VT had better survival than
12040343 underwent (defined those who did not after
revascularization as VT with syncope or the index event (HR: 0.67;
after a life- VT with symptoms and p=0.002). With a mean
threatening VA have LVEF 0.40 [VT/VF]). follow-up period of
improved survival Also, patients with 24.213.5 mo, crude
rate when unexplained syncope death rates (with 95%
compared with who had inducible and confidence limits) were
those who did not symptomatic VT during 21.4%4.8% in the
undergo revasc; and EPS. revascularization group
evaluate the and 29.4%2.0% in the
interaction of medically treated group.
revascularization
with ICD therapy After adjustment, HR
unchanged at 0.67,
Study type: significance decreased to
observational p=0.01.

Size: 3117 patients The association of better


with life-threatening survival with ICD was
VA, of whom 2321 consistent regardless of
(77%) had CAD and revascularization status
281 (17%)
underwent CABG
after the index
event
Mondsert et Aim: determine the Inclusion criteria: LVEF Intervention: Revascularization was not
al. 2016 (76) impact of 40%, first clinical coronary associated with
26806581 revascularization on sustained VA, without revascularization significantly lower rate of
recurrent VA or ACS recurrent VA or death
death (multivariable HR: 0.86;
95% CI 0.601.24, p=0.43)
Study type: regardless of whether
observational complete or incomplete
(HR: 0.65; 95% CI 0.25

38

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 274 patients, 1.69, p=0.37) or PCI or
mean follow-up 6.2 CABG (HR: 1.02; 95% CI
y 0.531.94, p=0.96). ICD
associated with
significantly lower
mortality (HR: 0.23; 95%
CI 0.09 0.55, p=0.001).
Ngaage et al. Aim: assess the Inclusion criteria: Intervention: CABG Perioperative mortality
2008 (77) outcomes in patients who was 6.5%, and 5 y survival
18355509 patients undergoing underwent CABG with rate was 88%,
CABG after ischemic preceding VT or VF comparable to patients
VT/VF (after MI, without prior VT/VF.
with exercise, with
CA)

Study type:
observational

Size: 93 patients
undergoing CABG
Every et al. Aim: estimate the Inclusion criteria: Significant association of
1992 (78) possible effect of OHCA survivors, CABG with lower risk of
1593036 CABG on the neurologically subsequent CA during
subsequent recovered, coronary follow-up RR: 0.48; 95%
outcome of patients disease, no prior CABG CI 0.240.97, p=0.04).
who have been or other Also, lower cardiac
resuscitated from revascularization mortality (RR: 0 .65; 95%
CA CI 0.391.10, p=0.10).

Study type:
observational

Size: 265 patients,


85 treated with
CABG, 180 medical
management,

39

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
van der Burg et Aim: determine Inclusion criteria: VA Intervention: N/A Patients with
al. 2003 (79) relation between CA survivors with CAD ischemic/viable
14530201 ischemia, viability, myocardium (N=73) were
scar tissue (and revascularized if possible.
revascularization), ICD in 112 (72%) patients.
and the incidence of 15 cardiac deaths
VA (and survival) in occurred and 42 (29%)
patients with CA and patients had recurrent
coronary disease VA. Patients with events
Study type: (death or recurrence)
observational exhibited more often a
severely depressed LVEF
Size: 153 patients, (30%), more extensive
follow-up up to 3 y scar tissue, and less
ischemic/viable
myocardium on perfusion
imaging and
less frequently
underwent
revascularization.

Multivariate analysis
identified extensive scar
tissue and LVEF 30% as
the only predictors of
death/recurrent VA
PROCAT Aim: assess Inclusion criteria: Intervention: At least 1 significant
Dumas et al. the effect of an patients with OHCA immediate PCI coronary lesion was
2010 (80) invasive strategy for with presumed cardiac found in 304 (70%)
20484098 patients with OHCA etiology and with patients, in 128 (96%) of
on hospital survival. coronary angiogram 134 patients with ST-
performed at admission segment elevation, and in
Study type: 176 (58%) of 301 patients
observational without ST-segment
elevation. Multivariable
Size: 435 patients analysis showed
treated with an successful coronary

40

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
immediate coronary angioplasty to be an
angiogram at independent predictor of
admission with survival, regardless of the
coronary post resuscitation ECG
angioplasty if pattern (OR: 2.06; 95% CI:
possible 1.163.66).
PROCAT II Aim: assess the Inclusion criteria: Intervention: At least 1 significant
registry association between patients with OHCA immediate PCI coronary lesion was
Dumas et al. early PCI and with presumed cardiac found in 403 of 695
2016 (81) favorable outcome etiology and with patients (58%). A PCI was
27131438 (cerebral coronary angiogram performed in 199 of 695
performance performed at admission (29%). A favorable
category 1 to 2 at outcome was observed in
discharge) 87 of 200 (43%) in
patients with PCI
Study type: compared with 164 of
observational 495 (33%) in patients
without PCI (p=0.02).
Size: 695 patients After adjustment, PCI was
treated with an associated with a better
immediate coronary outcome (adjusted OR:
angiogram at 1.80; 95% CI: 1.092.97,
admission without p=0.02).
ST elevation on
post-resuscitation
ECG
SYNTAX Aim: To show PCI is Inclusion criteria: Intervention: PCI 1 endpoint: rates of At 12 mo, the rates of death
Serruys et al. noninferior to CABG previously untreated with Taxus Express major adverse cardiac or and MI were similar between
2009 (82) for major adverse three-vessel or left paclitaxel-eluting cerebrovascular events at the 2 groups; stroke was
19228612 cardiac or main CAD (or both) stents 12 mo were significantly significantly more likely to
cerebrovascular with stable/unstable higher in the PCI group occur with CABG (2.2%, vs.
event (i.e., death angina or atypical chest Comparator: CABG (17.8%, vs. 12.4% for 0.6% with PCI; p=0.003).
from any cause, pain CABG; p=0.002)
stroke, MI,
or repeat Exclusion criteria:
revascularization)
during 12 mo

41

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Previous PCI or CABG,
Study type: RCT AMI, or the need for
concomitant cardiac
Size: 1800 patients surgery
with 12 mo follow-
up

SYNTAX Aim: to investigate Inclusion criteria: Intervention: PCI 1 endpoint: SCD: 24 (2.8%) with PCI, 15
Milojevic et al. specific causes of previously untreated 3- with Taxus Express 97 deaths after CABG and (1.9%) with CABG, HR: 1.61;
2016 (83) death, and its vessel or left paclitaxel-eluting 123 deaths after PCI 95% CI: 0.833.11, p=0.16.
26764065 predictors, after main CAD (or both) stents during a 5 y followup.
revascularization with stable/unstable After CABG, 49.4% of
for complex CAD in angina or atypical chest Comparator: CABG deaths were
patients pain cardiovascular, with the
greatest cause being
Study type: RCT Exclusion criteria: heart failure, arrhythmia,
Previous PCI or CABG, or other causes (24.6%).
Size: 1800 patients AMI, or the need for After PCI, the majority of
with 12 mo follow- concomitant cardiac deaths were
up surgery cardiovascular (67.5%)
and as a result of MI
(29.3%).
Treatment with PCI vs.
CABG was an
independent predictor of
cardiac death (HR: 1.55;
95% CI: 1.092.33; p =
0.045).
SCD-HeFT Aim: examine the Inclusion criteria: Intervention: ICD There was no significant
Al-Khatib et al. effect of the ICD on Overall SCD-HeFT, difference in ICD benefit
2008 (84) the outcomes of NYHA class II or III CHF Comparator: no ICD across the
18479330 patients with prior symptoms and a LVEF revascularization
coronary 35% due to ischemic subgroups (all p>0.1).
revascularization or nonischemic heart There was a trend toward
enrolled in SCD- disease. improved survival with an
HeFT ICD in patients who had

42

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
This substudy, patients their CABG >2 y before
Study type: RCT with ischemic heart randomization (HR: 0.71;
disease who were not 95% CI: 0.491.04) that
Size: of the 882 randomized to was not observed in
patients who met amiodarone (N= 884) patients who had their
these inclusion and who had complete CABG 2 y before
criteria, 255 (29%) revascularization data randomization (HR:1.40;
had no prior (revascularization 95% CI: 0.613.24)
revascularization, data were missing on 2
178 (20%) had prior patients).
PCI only, 284 (32%)
had prior CABG only,
and 165 (19%) had
prior PCI and CABG.
Nageh et al. Aim: assess the role Inclusion criteria: Overall group rates The 1 endpoint of total
2014 (85) of ICD in cardiac cardiac surgery and ICD mortality and appropriate
25146702 surgery patients within 3 mo shocks were observed in
with perioperative 52
resuscitated VA 35 (38%) and 28 (30%) of
arrest patients, respectively
<3 mo post
revascularization, Conclusion was that
and the role of ICDs recurrent VA are not
in prevented by CABG
patients who had
revascularization
after SCD

Study type:
observational,
evaluating total
mortality and/or
appropriate ICD
therapy

Size: 164 patients


had cardiac surgery

43

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
and ICD within 3
mo; 93/164 had an
ICD for sustained
pre- or
postoperative VT or
fibrillation requiring
resuscitation, mean
follow-up 49 mo

Data Supplement 12. Nonrandomized Trials, Observational Studies, and/or Registries of Arrhythmic Surgery and Revascularization for
Arrhythmia Management (Section 5.5.1)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Relevant 2 Endpoint (if any);
Author; Study Size (P values; OR or RR; Study Limitations;
Year Published & 95% CI) Adverse Events
Kumar et al. 2015 Aim: To characterized Inclusion criteria: Sixty- 1 endpoint: abolishment of The conclusion was that a
(86) the reasons for VT seven patients with VT at least 1 inducible VT, collaborative strategy of alternative
25925229 ablation failure and refractory to 42 AAD and complete success, partial interventional procedures offers the
describe alternative 21 previous success (abolishment of at possibility of achieving arrhythmia
interventional endocardial/epicardial least 1 spontaneous VT), and control in high-risk patients with VT that
procedures. catheter ablation failure (residual inducibility of is otherwise uncontrollable with AAD
attempts underwent spontaneous VT). and standard percutaneous catheter
Study type: Single transcoronary ethanol ablation techniques.
center experience ablation, surgical Results: Transcoronary
epicardial window (Epi- ethanol ablation alone was
Size: 62 window), or surgical attempted in 37 patients, OR-
cryoablation Cryo alone in 21 patients, and
a combination of
transcoronary ethanol
ablation and OR-Cryo (5
patients), or transcoronary
Exclusion criteria: N/A ethanol ablation and Epi-
window (4 patients), in the
remainder. Overall,
alternative interventional
procedures abolished 1
inducible VT and terminated

44

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
storm in 69% and 74% of
patients, respectively,
although 25% of patients had
at least 1 complication. By 6
mo post procedures, there
was a significant reduction in
ICD shocks (from a median of
8/mo to 1; p<0.001) and AAD
requirement although 55% of
patients had at least 1 VT
recurrence, and mortality was
17%.
Anter et al. 2011 (87) Aim: Evaluate the Inclusion criteria: Eight 1 endpoint: Clinical VT and The authors concluded that VT
21673018 efficacy of patients with recurrent ICD shocks circuits inaccessible to percutaneous
preoperative sustained VT refractory to ablation techniques are rare but can
electroanatomic and AAD underwent Results: During a mean be encountered in patients with
EP characterization of endocardial and/or followup period of 23 6 mo nonischemic cardiomyopathy. These
the VT substrate and epicardial ablation (range, 15 34 mo), 6 patients VTs can be successfully targeted by
circuit to guide procedures. After the had significant reduction in surgical cryoablation guided by
surgical ablation in unsuccessful VT burden as evident by a preoperative electroanatomic and
patients with NICM percutaneous approach, reduced number of ICD EP mapping.
surgical cryoablation was shocks after ablation (6.60.6
Study type: Single applied to the sites shocks per pt; p=0.026). Two
center experience previously identified and patients died, 1 of progressive
targeted during the HF and 1 of sepsis.
Size: 62 percutaneous procedure.

Exclusion criteria: N/A


Bhavani et al. 2007 Aim: To present Inclusion criteria: 3 1 endpoint: Successful Patient with intraoperatively CARTO
(88) variety of ablation patients who underwent elimination of VT
18039225 strategies and succeesful surgical
technologies for cryoablation after Results: Case report. The
surgical cryoablation catheter failed. specific approach
of VT (endocardial vs. epicardial,
Exclusion criteria: N/A beating heart vs. arrested)
and ablation device must be

45

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: Single tailored to the patient's
center experience- anatomy and presentation
case report

Size: 3
Sartipy et al. 2006 Aim: The aim of this Inclusion criteria: From 1 endpoint: Mortality and Vt Authors concluded that the Dor
(89) study was to evaluate July 1997 to December inducible or spontaneous procedure including endocardiectomy
16368337 the Dor procedure 2003, 53 consecutive and cryoablation yields a very high (90%)
including VT surgery patients with left Results: Early mortality was 2 freedom from spontaneous VT and
ventricular aneurysm and of 53 (3.8%). Mean followup eliminates the need for an ICD in most
Study type: Single VT underwent surgical was 3.7 y. At 1, 3, and 5 y patients
center experience ventricular restoration overall actuarial survival was Karolinska Institute is a specialized
including nonguided 94%, 80%, and 59%, center.
Size: 53 endocardiectomy and respectively. Surgical success
cryoablation. Twenty-four rate in patients with
patients had at least 1 preoperative spontaneous VT
preoperative episode of was 91%. Inducible VT was
spontaneous VT, and 29 found in 5 of 35 patients who
patients had inducible- underwent postoperative
only VT. programmed stimulation.
There was no arrhythmia-
Exclusion criteria: N/A related late death and there
was no loss to follow-up.
Choi et al. 2015 (90) Aim: The aim is to Inclusion criteria: During 1 endpoint: Patients The authors concluded that surgical
25697752 describe surgical the period from March outcomes. cryoablation is an option for highly
cryoablation of VA 2009 to March 2014, 190 symptomatic drug-resistant VAs
from the LVOT region consecutive patients with Results: Surgical cryoablation emanating from the LVOT region. Yet,
inaccessible for focal VA originating from was successful in 3 of the 4 the procedure is not effective for all
ablation because of the LVOT underwent patients. The 4th patient patients, and coronary injury is a risk.
epicardial fat or ablation at Brigham and subsequently had successful
overlying coronary Women's Hospital, endocardial catheter ablation.
arteries Boston. The study During a mean followup of 22
describes 4 patients (2%) 16 mo (range 442 mo), all
Study type: Single who underwent surgical patients showed abolition of
center experience cryoablation. or marked reduction in
symptomatic VA. However, 1
Size: 4 patient subsequently required

46

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
percutaneous intervention to
Exclusion criteria: N/A the LAD; another developed
progressive left ventricular
systolic dysfunction caused by
NICM; and a third patient
underwent permanent
pacemaker implantation
because of complete AV block
after concomitant aortic valve
replacement.
Patel et al. 2016 (91) Aim: to determine Inclusion criteria: From Endpoint: post LVAD VA. Open-chest hybrid epicardial mapping
26377813 effectiveness of hybrid March 2009 to October and ablation for recurrent VT is feasible
surgical epicardial 2012, 5 patients (4 men Results: Epicardial mapping and can be considered in select patients
mapping and ablation and 1 woman, age range was considered if patients during the period of LVAD implantation.
at the time to LVAD 5273 y) underwent open had recurrent VT despite
placement chest EPS and epicardial failed prior endocardial
mapping for recurrent VT ablation and/or
Study type: Single while the heart was electrocardiogram (EKG)
center experience. exposed during the features of an epicardial exit.
Retrospective review. period of LVAD Activation and/or a substrate
implantation mapping approach were
Size: 5 employed during all
Exclusion criteria: N/A procedures. 3 of 5 patients
(60%) had acute procedural
success. In all patients, VT
was either eliminated or
significantly reduced with
epicardial ablation. 1 patient
had mediastinal bleeding
delaying sternal closure.
During a follow-up period of
363368 d, 4 patients died
due to nonarrhythmic causes.
Mulloy et al. 2013 Aim: to determine Inclusion criteria: 50 1 endpoint: post LVAD Postoperative VA can be minimized by
(92) whether consecutive patients ventricualr arrhythmias. preoperative risk assessment and
22520722 intraoperative undergoing implantation intraoperative treatment. Localized
cryoablation in select of the HeartMate II LVAD cryoablation in select patients offers

47

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
patients reduces the were examined. 14 of Results: Compared with promising early feasibility when
incidence of these patients had NoCryo, the Cryo group had performed during HeartMate II LVAD
postoperative VA after recurrent preoperative significantly decreased implantation.
LVAD. VA. Of those patients with postoperative resource use None of the Cryo patients had
recurrent VA, half and complications (p<0.05). recurrent postoperative VA compared
Study type: Single underwent intraoperative Recurrent postoperative VA with 4 (57%) of the NoCryo group
center experience. cryoablation (Cryo: N=7) did not develop in any of the (p=0.02).
Retrospective review. and half did not (NoCryo: Cryo patients (p=0.02).
N=7).
Size: 14
Exclusion criteria: N/A

Data Supplement 13. RCTs for Autonomic Modulation (Section 5.6)


Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if
Author; Study Type; (# patients) / (Absolute Event Rates, any);
Year Published Study Size (N) Study Comparator P values; OR or RR; & Study Limitations;
(# patients) 95% CI) Adverse Events
Schwartz PJ et al. Study type: Inclusion criteria: Intervention: High risk: 1 endpoint: SCD. LCSD may be considered
1992 (93) RCT Patients post-MI (30 1:1:1 BB (oxprenolol) vs. 22 mo as a possible alternative for
d); High risk (evidence LCSD; High Risk: high-risk patients with
Aim: To explore the of Vfib or Vtach); low Low risk: BB vs. placebo. Placebo 21.3% contraindications to BB.
influence of BB vs. risk (no evidence of VF Oxprenolol 2.7%
LCSD in patients at or VT. Comparator: Placebo LCSD 3.6%
high risk for SCD.
Exclusion criteria Low Risk:
Size: 144 high risk; N/A Placebo: 5.2%
869 low risk Oxprenolol: 1.6%
Krittayaphong et al. Study type: Inclusion criteria: Intervention: 1 endpoint: BB may be useful for
2002 (94) RCT VA with LBBB, inferior Atenolol 50-100mg/day Atenolol significantly patients with RVOT and
12486439 axis morphology. decreased PVC count symptomatic VA.
Aim: To determine Symptomatic (VA Comparator: Placebo (p=0.001) and average
the efficacy of disturbed their daily heart rate (p<0.001)
atenolol in the activities) compared to placebo.
treatment of Both placebo and
symptomatic VA Exclusion criteria

48

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
from RVOT SHD. atenolol decreased
compared with symptom frequency.
placebo

Size: 52

49

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 14. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries for Autonomic Modulation (Section 5.6)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Vaseghi et al. 2014 Study type: Inclusion criteria: 1 endpoint: Survival free of ICD Bilateral cardiac sympathetic
(95) retrospective chart VT storm (>3 events shocks. denervation appears better than
24291775 review requiring treatment in 24 LCSD
h) or refractory VA and Results:
Aim: To describe the ICD shocks who Survival free of ICD shocks: 30%
experiences of underwent cardiac in LCSD; 48% in the BCSD.
patients with VT storm sympathetic denervation (p=0.04)
who underwent between April 2009 and
cardiac sympathetic December 2012. number of shocks decrease from
denervation. Mean of 19 pre CSD to 2.3
Exclusion criteria: (p<0.001)
Size: N= 41 (14 LCSD; N/A
27 BCSD)
Ajijola et al. 2012 (96) Study type: Case Inclusion criteria: 1 endpoint: Reduction in Ventricular Our study suggests that
22192676 Series Patients with ongoing events patients with incessant VA for
VAs with LCSD and whom no other therapeutic
Aim: To describe the maximal med therapy Results: options exist, bilateral cardiac
experiences of Complete response in 4/6 sympathetic denervation may be
patients with bilateral Exclusion criteria: N/A Partial response in 1/6 beneficial.
cardiac sympathetic No response in 1/6 (PMVT)
denervation (or RCSD
after unsuccessful
LCSD)

Size: N=6
Ukena et al. (97) Study type: Inclusion criteria: 1 endpoint: Reduction in Ventricular Renal sympathetic denervation
27364940 Multicenter (5) Case CHF; Recurrent VA events appeared safe and was
Series refractory to medications associated with a reduction in
and ablation Results: VT/VF events.
Aim: To describe the Median VT/VF:
effect of renal Exclusion criteria: N/A 4 wk prior =21
denervation on 1 mo post =2 (p=0.004)
refractory VT 3 mo post =0 (p=0.006)
50

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: N=13 No peri-procedural adverse events
Baseline BP was low but no change in
BP.
Grimaldi et al. 2012 Study type: Case Inclusion criteria: 1 endpoint: Ventricular arrhythmia SCS may decrease the rate of
(98) Series (from patients Patients with CM, ICDs VA.
22877745 enrolled in an under- and previous VF or 2xVT Results:
enrolled RCT trial Patient 1 had a 75% reduction in VA
was a 2 mo alternating Exclusion criteria: N/A with SCS on
on/off design.) Patient 2 had a 100% reduction in VA
with SCS on.
Aim: To describe the (These are the authors reports,
experiences of numbers in the table dont quite add
patients with SCS on to this. Not sure how the math was
done)
Size: N=2

Data Supplement 15. RCTs Comparing Acute Management of Specific Arrythmias - (Section 6)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if any);
Author; Study Type; (# patients) / (Absolute Event Rates, Study Limitations;
Year Published Study Size (N) Study Comparator P values; OR or RR; & Adverse Events
(# patients) 95% CI)
Kudenchuk et Aim: Compare Inclusion criteria: 18 y Intervention: IV 1 endpoint: No Neurologic outcomes similar
al. 2016 (99) amiodarone, or older with OHCA and amiodarone or difference in survival to More amiodarone patients
27043165 lidocaine, placebo in shock refractory VF or lidocaine; repeated hospital discharge: required temporary pacing;
OHCA with shock- pulseless VT. IV access once if VF/VT amiodarone (24.4%), otherwise, no difference in
refractory VF or persisted after initial lidocaine (23.7%), drug related adverse events
pulseless VT Exclusion criteria: dose and repeat placebo (21.0%). Trial may have been
Already received shocks Amiodarone vs. placebo underpowered to show
Study type: RCT lidocaine or 3.2% points (95% CI: -0.4 amiodarone benefit over
double-blind, amiodarone, Comparator: IV 7.0; p=0.08); lidocaine vs. placebo
placebo controlled hypersensitivity to normal saline placebo 2.6% points (95%
these drugs repeated once if CI: -1.06.3; p=0.16); Note: An editorial (100)
Size: 3,026 patients VF/VT persisted after Amiodarone vs. lidocaine suggesting use of amiodarone

51

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
initial dose and 0.7% points (95% CI: -3.2 or lidocaine for witnessed
repeat shocks 4.7; p=0.70) arrest as there was a significant
reduction in shocks and fewer
In witnessed arrest, CPR events in hospital.
survival to hospital
discharge with
amiodarone and lidocaine
was higher than with
placebo. The absolute risk
difference for
amiodarone vs. placebo
was (5.0 % points, p=0.04)
and for lidocaine vs.
placebo was (5.2 %
points, p=0.05)
Jacobs et al. Aim: Compare Inclusion criteria: Age Intervention: 1 ml 1 endpoint: Survival to Epinephrine improved return
2011 (101) epinephrine with 18 y with OHCA, CPR aliquots of hospital discharge not to spontaneous circulation but
21745533 normal saline during started by paramedics epinephrine 1:1000 different: 1.9% for not survival to hospital
OHCA treated following current placebo and 4% for discharge
following ACLS Exclusion criteria: ACLS guidelines epinephrine (OR: 2.2; 95% Limitations: Inadequate
guidelines Traumatic OHCA CI: 0.76.3). Return of sample size to access hospital
Comparator: 1 ml spontaneous circulation survival.
Study type: RCT aliquots of 0.9% 8.4% for placebo and Quality of ACLS not
double blind, sodium chloride 23.5% for epinephrine evaluated
placebo controlled following current (OR: 3.4; 95% CI: 2.05.6) Adverse events not listed
ACLS guidelines
Size: 601 patients

52

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Piccini et al. Aim: Compare Inclusion criteria: Intervention: BB 1 endpoint: BB therapy Sustained VT/VF was a major
2008 (102) outcomes in acute MI with sustained within 24 h of MI within 24 h was predictor of in-hospital death
19026290 patients with MI and VT/VF and/or high Killip associated with (RR: 4.18; 95% CI: 2.915.93)
sustained VT/VF classification Comparator: No BB decreased in-hospital
treated or not mortality in patients with
treated with BB Exclusion criteria: N/A sustained VT/VF (RR:
0.28; 95% CI: 0.100.75,
Study type: p=0.013) without
Prospective, evidence of worsening HF
multicenter registry 55.2% of patients with
of patients with sustained VT/VF were
acute MI treated with BB within 24
h of MI
Size: 306 patients
with sustained
VT/VF
Dorian et al. Aim: Compare IV Inclusion criteria: Age Intervention: 1 endpoint: Amiodarone Increased survival with
2002 (103) lidocaine with IV 18 y with OHCA due to Patients randomized had higher survival to shorter interval from dispatch
11907287 amiodarone as VF. to IV amiodarone hospital admission than to receiving study drugs.
adjunct to plus IV lidocaine lidocaine: 28% with Patients with VF had better
defibrillation in Exclusion criteria: placebo or IV amiodarone vs. 12% with survival than those with
OHCA traumatic, or OHCA lidocaine plus IV lidocaine (OR: 2.17; 95% asystole or PEA.
amiodarone placebo CI: 1.213.83; p=0.009). Amiodarone did not improve
Study type: RCT to treat VF resistant Of 42 patients surviving survival to hospital discharge
placebo controlled to 3 shocks, at least 1 to hospital admission, 9 Limitation: not powered to
dose of IV (5%) survived to hospital show amiodarone improved
Size: 347 patients epinephrine, and discharge in the survival to discharge.
then 4th shock. Or, amiodarone group and of No adverse events noted.
recurrent VF after 20 initial survivors in the
successful initial lidocaine group, 5 (3%)
shock. were discharged (p=0.34).

Comparator: 1 ml
aliquots of 0.9%
sodium chloride
following current
ACLS guidelines
53

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Hassan et al. Aim: IV magnesium Inclusion criteria: Intervention: 1 endpoint: IV No benefit from magnesium
2002 (104) given early during Patients 18 y with Patients received 24 magnesium did not Limitations: Possible
11777881 CPR for VF will OHCA and refractory or g of magnesium improve survival to inadequate magnesium dose
improve survival. recurrent VF hospital admission: 17% No adverse effects listed
Comparator: for magnesium and 13%
Study type: RCT, Exclusion criteria: Placebo for placebo (OR: 1.69;
double blind, Traumatic OHCA 95% CI: -10%18%)
placebo controlled

Size: 105 patients


MAGIC Aim: Determine if Inclusion criteria: Intervention: IV 1 endpoint: Magnesium No benefit of magnesium for
Thel et al. 1997 IV magnesium Adult patients with CA magnesium bolus did not improve return to survival to 24 h or hospital
(105) improves return to in the ICU or hospital followed by a 24 h spontaneous circulation: discharge
9357406 spontaneous wards infusion 54% with magnesium and No adverse effects
circulation 60% with placebo (95%
(measurable BP and Exclusion criteria: Comparator: Normal CI: 0.410.47; p=0.44)
pulse) for 1 h after Patients in emergency saline
in-hospital CA department. Advanced
heart block, chronic
Study type: RCT, renal failure, already on
placebo controlled magnesium

Size: 156 patients


Somberg et al. Aim: Establish the Inclusion criteria: Intervention: IV 1 endpoint: Amiodarone was more
2002 (106) effectiveness of IV Patients with incessant amiodarone (or IV Amiodarone was more effective than lidocaine for
12372573 amiodarone for (shock resistant) VT not lidocaine) followed effective than lidocaine: terminating VT with improved
shock resistant VT. treated with prior by a 24 h infusion. If amiodarone terminated 24 h survival.
antiarrhythmics the first medication VT in 78% and lidocaine Limitations: Drug related
Study type: RCT, failed to terminate 27% (p<0.01). OR and CI hypotension with amiodarone
double-blinded, Exclusion criteria: VT, patients were not listed. 24 h survival less frequent than with
parallel design Already on AAD crossed over to the 39% on amiodarone and lidocaine.
alternative 9% on lidocaine (p<0.01).
Size: 29 patients medication. More hypotension with
lidocaine than
Comparator: amiodarone (28% vs. 7%,
Lidocaine

54

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
p=0.06). Bradycardia
equal

Kudenchuk et Aim: Determine if Inclusion criteria: Intervention: IV 1 endpoint: Amiodarone Amiodarone improved
al. 1999 (107) amiodarone Patients <18 with OHCA amiodarone (single improved survival to survival to hospital with no
10486418 improves the rate of due to VF or pulseless dose) after receiving hospital admission: 44% difference in duration of
successful VT that remained 1 mg epinephrine on amiodarone and 34% resuscitation, number of
resuscitation after present after 3 shocks, on placebo (OR: 1.6; 95% shocks, need for other
OHCA with IV access Comparator: CI: 1.12.4; p=0.02) antiarrhythmics
Placebo (polysorbate Limitations: lack for power to
Study type: RCT, Exclusion criteria: 80, dilutant, single detect treatment effect on
double blinded, Absence of IV access, dose) after receiving survival to hospital discharge
placebo controlled VF, or pulseless VT 1 mg epinephrine More hypotension with
amiodarone (59% vs. 48%,
Size: 504 patients p=0.04)

Callaham et al. Aim: To determine Inclusion criteria: Intervention: High 1 endpoint: High dose High dose epinephrine
1992 (108) the relative efficacy Adults with OHCA who dose epinephrine (15 epinephrine significantly improved admission to hospital
1433686 of high vs. standard would receive mg), high dose improved the rate of but no difference in dismissal
dose epinephrine by AHA norepinephrine (11 return of spontaneous from hospital
catecholamines in ACLS guidelines mg), or standard circulation: 13% for high Trends for norepinephrine
initial treatment of dose epinephrine dose epinephrine, 8% were not different
OHCA Exclusion criteria: blindly substituted receiving standard dose Limitations: low hospital
None listed for ACLS doses of epinephrine (p=0.01). dismissal rate
Study type: RCT, epinephrine 18% of high dose No adverse effects
double blind epinephrine and 10% of
Comparator: standard dose
Size: 816 patients standard dose epinephrine patients
epinephrine (no admitted to hospital
placebo) (p=0.02)

55

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Gueugniaud et Aim: compare Inclusion criteria: Intervention: High 1 endpoint: 40.4% of Long-term survival after
al. 1998 (109) repeated low dose OHCA patients with dose epinephrine, 5 1677 patients in the high OHCA was no better with
9828247 vs high dose VF/VT despite mg, up to 15 doses dose group had a return repeated high doses of
epinephrine in defibrillation shocks, or of spontaneous epinephrine than with
OHCA asystole /hypotensive Comparator: circulation compared to repeated standard doses.
VT standard dose 36.4% of 1650 patients in
Study type: epinephrine, 1 mg, the standard dose group
Prospective, Exclusion criteria: following ACLS (p=0.02). There was no
multicenter, Inadequate data protocol difference in survival to
randomized hospital discharge (2.3%
vs 2.8%. p=0.34).
Size: 3327 patients
Gorgels et al. Aim: Determine the Inclusion criteria: Intervention: IV 1 endpoint: Procainamide was superior
1996 (110) relative efficacy of Adult patients with procainamide (10 Procainamide was more to lidocaine for terminating VT
8712116 procainamide and spontaneous mg/kg at 100 effective than lidocaine: Limitations: No patients with
lidocaine for monomorphic VT mg/min) or lidocaine 27% of VT episodes AMI or ischemia
treating (1.5 mg/kg over 2 responded to lidocaine Significant lengthening of
spontaneous Exclusion criteria: min) and 77% to procainamide QRS and QT on procainamide
monomorphic VT Patients with AMI and (p<0.01)
those with poor Comparator:
Study type: hemodynamic Procainamide or
Randomized, open tolerance lidocaine (no
label, parallel study placebo)

Size: 29 patients
Ho et al. 1994 Aim: Determine the Inclusion criteria: Intervention: IV 1 endpoint: Sotalol was No 2 endpoints
(111) relative efficacy of Adult patients with sotalol (100 mg) more effective than Limitations: no placebo
7912296 lidocaine and sotalol sustained VT lidocaine for terminating control; small number of
for terminating Comparator: IV VT: 69% with sotalol and patients
spontaneous VT not Exclusion criteria: lidocaine (100 mg) 18% with lidocaine (95% 1 death in each drug group
causing CA Already on an CI: 22%80%; p=0.003) after the first drug and 1 death
antiarrhythmic, Cross-over to second in each group after both drugs
Study type: RCT, hypotension requiring drug if VT persisted
double blind immediate after 15 min
cardioversion, known
Size: 33 patients adverse reaction to
either medicantion

56

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Levine et al., Aim: Response rate Inclusion criteria: Intervention: 1 endpoint: 110 patients Significantly longer time to
1996 (112) and safety Patients with recurrent Patients (40.3%) survived 24 h first recurrence in the 2 higher
8522712 of intravenous hypotensive VT were randomized to without another dose groups
amiodarone in refractory to lidocaine, receive 1 of 3 doses hypotensive VT episode Hypotension required
patients with VT procainamide and of intravenous vasopressor therapy in 38
refractory to bretylium. amiodarone: 525, Safety endpoint: Adverse patients (14%) and led to death
standard therapies. 1,050 or 2,100 mg/24 events requiring drug in 6 (2%).
Exclusion criteria: h by continuous discontinuation
Study type: Cardiogenic shock; infusion over 24 h.
prospective, significant hepatic
controlled dysfunction or Comparator: As
pulmonary disease; Hx above
Size: 273 patients of TdP; congenital QT
prolongation;
bradyarrhythmias or AV
block (unless
pacemaker present).
Teo et al. 1993 Aim: Assess the Inclusion criteria: Intervention: AAD 1 endpoint: 660 deaths The routine use of Class I
(113) effectiveness of AAD Patients with AMI in 11,712 patients agents (lidocaine,
8371471 on mortality in randomized to AAD Comparator: receiving Class I agents procainamide) was associated
patients with AMI therapy Placebo, standard and 571 deaths in 11,517 with increased mortality after
agents controls (OR: 1.14; 95% MI.
Study type: Exclusion criteria: CI: 1.011.28; p=0.03). BB reduced morality
Metanalysis Inadequate study 778 patients received The amiodarone data was
design amiodarone and 77 died, limited but promising
Size: 138 compared with 101
randomized trials, deaths in 779 control
98,000 patients patients (OR, 0.71; 95%
CI, 0.510.97, p=0.03).
26,973 patients received
BB and 1,464 died
compared with 1,727
deaths in 26,295 controls
(OR: 0.81; 95% CI, 0.75
0.87, p=0.00001)

57

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Elzari et al. Aim: Assess the Inclusion criteria: Intervention: IV or 1 endpoint: The study Amiodarone given by IV and
2000 (114) mortality associated Acute MI, no PO amiodarone was modified after the PO to a total of 2,700 mg in the
10639301 with amiodarone in contraindications to first 516 patients showed first 48 h after MI was
patients with AMI study drug Comparator: Placebo higher mortality on associated with increased
amiodarone than placebo mortality.
Study type: Single (16.30% vs. 10.16%; Reducing the dose by half
center, randomized p=0.04). showed amiodarone and
Exclusion criteria: placebo mortality were similar
Size: 1,073 patients Contraindication to Safety endpoint:
amiodarone Increased mortality on
high dose amiodarone

Data Supplement 16. Nonrandomized Trials, Observational Studies, and/or Registries Comparing Acute Management of Specific Arrythmias
(Section 6)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Piccini et al. 2008 Study type: Registry of Inclusion criteria: 1 endpoint: death Sustained VT/VF was common with AMI
(102) patients in the Patients with AMI who Results 306 of 5,391 patients In patients with sustained VT/VF, BB
19026290 VALsartan In Acute experienced sustained (5.7%) in the VALIANT registry therapy in the first 24 h after AMI was
myocardial iNfarcTion VT/VF had sustained VT/VF with a associated with decreased early mortality
trial (VALIANT) Exclusion criteria: mortality of 20.3%. 55.2% without worsening HF.
inadequate data were treated with IV or oral
Size: 306 patients BB which were associated
with decreased in-hospital
mortality (RR: 0.28; 95% CI:
0.100.75, p=0.013)
Link et al 2015 (115) Study type: Guidelines Inclusion criteria: Acute Expert developed guidelines Electrical cardioversion is
26472995 treatment of patients recommended for the initial treatment of
with VA Reviews role of direct current VF, poorly tolerated VT, and polymorphic
cardioversion, epinephrine, VT.
magnesium, and AAD therapy The appropriate use of AAD,
for the treatment of acute VA epinephrine, and magnesium for the
treatment of acute VA is discussed
58

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Herlitz et al.1997 Study type: Inclusion criteria: All 1 endpoint: Survival to Lidocaine improved the return to
(116) Retrospective, patients with OHCA due hospital discharge spontaneous circulation and
9044490 observational study of to VF. CPR by single hospitalization
patients with OHCA center emergency Results: Patients receiving Lidocaine did not improve rate of
due to VF department lidocaine had a higher return discharge from hospital
of spontaneous circulation
Size: 1,212 cases; 405 Exclusion criteria: (p<0.001) and hospitalized
receiving lidocaine Traumatic cause of OHCA alive (38% vs. 18%; p<0.01).
Survival to discharge did not
differ
Markel et al. 2010 Study type: Inclusion criteria: 1 endpoint: The association Procainamide associated with more
(117) Retrospective, Witnesses, OHCA due to between procainamide and shocks, pharmacologic interventions, and
20624142 observational, cohort VF or pulseless VT survival longer resuscitations.
treated by King County, Procainamide did not improve survival
Size: 665 patients, WA, emergency services. Results: Procainamide
176 received associated with a lower
procainamide Exclusion criteria: survival to hospital discharge
Traumatic cause of (OR: 0.52; 95% CI: 0.360.75)
OHCA, asystolic OHCA
Stiell et al. 2004 Study type: Inclusion criteria: OHCA 1 endpoint: survival to The addition of ACLS did not improve
(118) Multicenter, hospital admission and the rate of survival over the use of rapid
15306666 controlled prospective Exclusion criteria: discharge defibrillation in OHCA.
trial traumatic cause of SCD
Results: The rate of hospital
Size: 5638 patients; admission increased from the
1391 enrolled in the defibrillation phase to the
rapid defibrillation ACLS phase (10.9% vs 14.6%,
phase and 4247 in the p<0.001). Survival after rapid
ACLS phase defibrillation (OR: 3.4; 95% CI:
1.48.4) was better than ACLS
(OR: 1.1; 95% CI: 0.81.5) and
bystander CPR (OR: 3.7; 95%
CI: 2.55.4)
Haqihara et al. 2012 Study type: Inclusion criteria: Age 1 endpoint: Return of Pre-hospital epinephrine for OHCA was
(119) Prospective, 18 y with OHCA treated spontaneous circulation, associated with improved return to
22436956 observational by emergence medical survival at 1 mo, neurologic spontaneous circulation.
service personnel outcome

59

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 417,188 patients Pre-hospital epinephrine for OHCA was
Exclusion criteria: Results: Epinephrine associated with worse 1 mo survival and
Traumatic cause of OHCA improved return of neurologic outcomes.
spontaneous circulation (OR:
2.36; 95% CI: 2.222.50;
p<0.001); but had an adverse
effect on long-term outcome
measures (1 mo survival, OR:
0.46; 95% CI: 0.420.51; and
neurologic, OR: 0.31; 95% CI:
0.260.36)
Donnino et al. 2014 Study type: Inclusion criteria: Adults 1 endpoint: Survival to Patients with non-shockable CA in
(120) Prospective data with CA in hospital with hospital discharge hospital had improved return of
24846323 collection, asystole or pulseless VT spontaneous circulation, survival in
observational as the initial rhythm Results: Survival was hospital, and neurologically intact
increased by early survival with earlier administration of
Size: 25,095 patients Exclusion criteria: administration of epinephrine
Cardiac arrest in epinephrine: 13 min
emergency department, (reference group) (OR: 1.0);
ICU, missing data, 46 min (OR: 0.91; 95% CI:
received vasopressin 0.821.0; p=0.055); 79 min
(OR: 0.63; 95% CI: 0.520.76;
p<0.001).
Koscik et al. 2013 Study type: Inclusion criteria: Adults 1 endpoint: Does timing of Early administration of epinephrine
(121) Retrospective with OHCA epinephrine administration improved return of spontaneous
23523823 database analysis improve outcome circulation
Exclusion criteria: Early administration of epinephrine did
Size: 686 patients Traumatic cause of OHCA Results: Early epinephrine not increase survival to admission or
was more likely to have discharge
return of spontaneous Similar results were reported with PEA
circulation (32% vs. 23.4%;
OR: 1.59; 95% CI: 1.072.38)
Spaulding et al. 1997 Study type: Inclusion criteria: OHCA 1 endpoint: Incidence of Acute coronary occlusion is frequent in
(122) Retrospective, survival acute coronary occlusion and survivors of OHCA and is predicted poorly
9171064 observational, role of reperfusion therapy by clinical and ECG findings
consecutive patients Exclusion criteria: Non- Coronary angioplasty may improve
cardiac cause of arrest survival

60

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 84 patients Results: 71% had significant
CAD and 48% had coronary
artery occlusion. In-hospital
survival 38%. Successful
angioplasty predicted survival
(OR: 5.2; 95% CI: 1.124.5;
p=0.04)
Cronier et al. 2011 Study type: Inclusion criteria: OHCA 1 endpoint: Prognostic Routine coronary angiography with
(123) Retrospective, survivor, age <80 y, impact of routine PCI percutaneous intervention may improve
21569361 observational, treated with mild survival following OHCA in patients
consecutive patients hypothermia, Results: 73% had CAD. Time treated with mild hypothermia who are
hemodynamically stable from collapse to return of hemodynamically stable
Size: 111 patients spontaneous circulation
Exclusion criteria: Non- associated with mortality (OR:
cardiac cause of arrest 1.05; 25th75tth percentile
range, 1.031.08; p<0.001);
Percutaneous intervention
associated with survival (OR:
0.30; 25th75th percentile
range, 0.110.79; p=0.01)
Zanuttini et al. 2012 Study type: Inclusion criteria: OHCA 1 endpoint: Independent Emergency coronary angiography and
(124) Retrospective, survival, remained determinants of in-hospital PCI, if indicated, appeared to improve
22975468 observational, unconscious soon after survival survival.
consecutive patients recovery of spontaneous The study has significant limitations: no
circulation Results: Coronary control group; and unconscious patients
Size: 93 patients angiography performed in 66 who had delayed procedures 18 d after
Exclusion criteria: Non- patients (71%); 48 emergent OHCA is a poor comparative group.
cardiac cause of OHCA and 18 at 1310 d. PCI in
52%; in hospital survival 54%.
Emergency angiography (HR:
2.32; 95% CI: 1.234.38;
p=0.009) and PCI (HR: 2.54;
95% CI: 1.354.8; p=0.004)
related to in hospital survival
Dumas et al. 2016 Study type: Inclusion criteria: OHCA 1 endpoint: Favorable 1/3 of OHCA patients without ST
(81) Observational, survivor without an ST- neurologic outcome elevation had a culprit lesion and had a
27131438 multicenter registry elevation MI

61

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Results: 199 patients (29%) nearly 2-fold increase in favorable
Size: 695 patients Exclusion criteria: had a PCI. 43% with PCI had a neurologic outcome.
Inadequate data favorable outcome and 33% A favorable outcome was also
without PCI. (OR: 1.80; 95% predicted by a shockable rhythm, lower
CI: 1.092.97; p=0.02). epinephrine dose, and shorter
resuscitation.

Kudenchuk et al. Study type: Inclusion criteria: OHCA 1 endpoint: re-arrest, Patients receiving lidocaine had a
2013 (125) retrospective, cohort due to VF or VT. Age 18 hospital admission, survival shorter time to a return of spontaneous
23743237 of patients with OHCA y circulation and higher BP
who did or did not Results: 1296 patients Use of prophylactic lidocaine upon
receive prophylactic Exclusion criteria: received prophylactic return to a spontaneous circulation after
lidocaine Missing data points, no lidocaine and 425 did not. OHCA was associated with less recurrent
chance of survival when Prophylactic lidocaine VF/VT and higher rates of admission to
Size: 1721 patients paramedics arrived reduced re-arrest from VF/VT hospital and survival to discharge.
with OHCA due to VF (OR: 0.34; 95% CI: 0.260.44);
or VT non-shockable arrhythmias
(OR: 0.47;95% CI: 0.290.78);
higher hospital admission
(OR: 1.88;95% CI, 1.282.76);
and improved survival to
discharge (OR, 1.49;95% CI:
1.151.95)
Nademanee et al., Study type: Inclusion criteria: ES 1 endpoint: Effect of beta Sympathetic blockade is superior to
2000 (126) retrospective, with recent (72 h3 mo) blockade (left stellate standard ACLS therapy in treating ES
10942741 observational MI ganglion blockade, esmolol, patients.
propranolol) on outcome
Size: 49 patients Exclusion criteria: MI <72 (survival)
h
Results: 1-wk mortality rate
was higher in group not
treated with beta blockade:
18 (82%) of the 22 patients
died, all of refractory VF,
compared to 6 (22%) of the
27 patients with beta
blockade, 3 of refractory VF

62

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
(p<0.0001). Patients who
survived the initial ES event
did well over the 1 y followup
period: Overall survival was
67% with beta blockade
compared with 5% without it
(p<0.0001).
Sasson et al. 2010 Study type: Meta- Inclusion criteria: OHCA 1 endpoint: survival Witnessed OHCA and arrest due to
(127) analysis OF OHCA VF/VT treated with defibrillation had
20123673 studies Results: Survival to hospital improved survival.
discharge was more likely
Size: 79 studies among OHCA patients
reporting 142,740 witnessed by a bystander
patients (6.4% to 13.5%); witnessed by
EMS (4.9% to 18.2%),
received bystander CPR (3.9%
to 16.1%), or were found in
VF/VT (14.8% to 23%).
Buxton et al 1987 Study type: single Inclusion criteria: 1 endpoint: adverse IV verapamil should not be used in
(128) center, observational Sustained VT treated hemodynamics patients with sustained VT
3578051 with IV verapamil
Results: 44% of 25 patients
with sustained VT receiving IV
Size: 25 patients verapamil had severe
hypotension of loss of
consciousness.
Pellis et al. 2009 Study type: Inclusion criteria: OHCA 1 endpoint: return of A pre-cordial thump did not delay other
(129) prospective, spontaneous circulation and aspects of CPR and had no adverse
19010581 observational Exclusion criteria: hospital discharge effects; but efficacy was lacking.
Inadequate data
Size: 144 patients Results: Precordial thump
had no effect on heart
rhythm in 96% of patients.
with return of spontaneous
circulation in only 3 patients.

63

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Volkman et al. 1990 Study type: single Inclusion criteria: 1 endpoint: VT conversion A pre-cordial thump converted VT in 77%
(130) center, observational, patients with VT following a pre-cordial thump of patients with a rate 160 bpm but only
2087859 consecutive patients 20% if the rate was faster. VF and VFL
Results: VT with a heart rate did not convert.
160 BPM converted in 17 of
22 cases, and VT >160 bpm
Size: 47 patients converted in 3 of 15 cases. 3
cases of VF and 7 cases of VFL
failed to convert.

Data Supplement 17. RCTs Secondary Prevention Sudden Death in Ischemic Heart Disease (Section 7.1.1)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint and Results Relevant 2 Endpoint (if any);
Author; Study Type; (# patients) / Study Limitations;
Year Published Study Size (N) Study Comparator Adverse Events
(# patients)
AVID Aim: To examine Inclusion criteria: Intervention: 1 endpoint: Study terminated early after
The AVID the effect on patients who were Therapy with ICD Overall survival was 1016 of 1200 patients enrolled
Investigators overall survival of resuscitated from greater with the ICD, with 81% of patients had CAD
1997 (131) initial therapy with near-fatal VF; Comparator: unadjusted estimates of Conclusion: Among survivors of
9411221 an ICD as sustained VT with Antiarrhythmic 89.3% as compared with VF or sustained VT causing severe
compared with syncope; or sustained drugs - amiodarone 82.3% in the symptoms, ICD is superior to AAD
amiodarone or VT with an LVEF 0.40 or sotalol, (only 2.6% antiarrhythmic-drug therapy for reducing overall
sotalol in patients and symptoms received sotalol) group at 1 y, 81.6% vs. mortality.
resuscitated from suggesting severe 74.7% at 2 y, and 75.4%
VF or symptomatic, hemodynamic vs. 64.1% at 3 y (p<0.02).
sustained VT with compromise. The corresponding
hemodynamic Exclusion criteria: reductions in mortality
compromise. arrhythmia was (with 95% CI) with the ICD
judged to have a were 3920%, 2721%,
Study type: RCT transient or and 3121%
correctable cause,
Size: 1016 excessively high risk
patients (life expectancy <1 y,
class IV HF, awaiting a
heart transplant, or

64

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
requiring a balloon
pump, other
mechanical means, or
inotropic drug
administration for
hemodynamic
support)
or excessively low risk
(event occurring
within 5 d of cardiac
surgery or
angioplasty, or
occurring in-hospital
<5 d after MI),
previous
ICD implant (or
attempted implant),
chronic serious
bacterial infection, or
were unable to give
verbal
assent due to
neurologic
impairment, or a
contraindication to
amiodarone
CIDS Aim: To compare Inclusion criteria: in Intervention: ICD 1 endpoint: Death from 82% had ischemic etiology
Conolly et al. the efficacy of the the absence of either any cause. Conclusions: CIDS provides
2000 (132) ICD and recent AMI or Comparator: A nonsignificant reduction further support for the superiority
10725290 amiodarone for the electrolyte imbalance, Amiodarone in the risk of death was of the ICD over amiodarone in the
prevention of they manifested any observed with the ICD, treatment of patients with
death in patients of the following: (1) from 10.2%/y to 8.3%/y symptomatic sustained VT or
with previous documented VF; (2) (RRR 19.7%; 95% CI: - resuscitated CA.
sustained VA OHCA requiring 7.7%40%; p=0.142). A
defibrillation or nonsignificant reduction
Study type: RCT cardioversion; (3) in the risk of arrhythmic
documented, death was observed, from

65

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 659 patients sustained VT causing 4.5%/y to 3.0%/y (RRR
syncope; (4) other 32.8%; 95% CI, -7.2%
documented, 57.8%; p=0.094).
sustained VT at a rate
150 beats/min,
causing presyncope or
angina in a patient
with a LVEF 35%; or
(5) unmonitored
syncope with
subsequent
documentation of
either spontaneous
VT10 s or sustained
(30 s) monomorphic
VT induced by
programmed
ventricular
stimulation.
Exclusion criteria: (1)
ICD or amiodarone
not considered
appropriate, (2)
excessive
perioperative risk for
ICD implantation; (3)
previous amiodarone
therapy for 6 wk; (4)
nonarrhythmic
medical condition
making 1y survival
unlikely, and (5) long-
QT syndrome.

66

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
CASH Aim: to study the Inclusion criteria: Intervention: ICD 1 endpoint: The 1 end In ICD patients, the percent
Kuck et al. impact on overall patients resuscitated therapy point was all-cause reductions in all-cause mortality
2000 (133) survival of initial from CA 2 to mortality. were 41.9%, 39.3%, 28.4%, 27.7%,
10942742 therapy with an documented Comparator: Over a mean followup of 22.8%, 11.4%, 9.1%, 10.6%, and
ICD as compared sustained VA amiodarone, 5734 mo, the death 24.7% at y 1 to 9 of followup.
with that with 3 metoprolol, or rates were 36.4% (95% CI Coronary disease was etiology in
AAD Exclusion criteria: If propafenone. 26.9% to 46.6%) in the 73%. A much larger reduction of
CA occurred within 72 Assignment to ICD and 44.4% (95% CI 61%, for SCD was observed
Study type: RCT h of an AMI, cardiac propafenone was in 37.2% to 51.8%) in the
surgery, electrolyte March 1992, after an amiodarone/metoprolol
Size: 288 patients abnormalities, or interim analysis arm. Overall survival was
proarrhythmic drug showed a 61% higher, though not
effect. higher all-cause significantly, in patients
mortality rate than assigned to ICD than in
in 61 ICD patients those assigned to drug
during a followup of therapy (1-sided p=0.081,
11.3 mo. HR: 0.766; 97.5% CI upper
bound 1.112)
Connolly et al. Aim: To obtain the Inclusion criteria: Intervention: 1 endpoint: Reduction 2 endpoints:
2000 (134) most precise RCTs ICD (934 patients) in death from any cause Arrhythmic death, HR 0.50 (95% CI
11102258 estimate of the evaluating the ICD vs. with the ICD, HR 0.72; 0.37-0.67; p<0.0001).
efficacy of the ICD, AAD therapy in Comparator: 95% CI 0.60-0.87; Survival was extended by a mean of
compared to patients with Amiodarone (932 p=0.0006). 4.4 mo by the ICD over a followup
amiodarone, for sustained VA or SCD patients) period of 6 y.
survival in patients P heterogeneity=0.306
with malignant VA. Patients with LVEF 35% derived
more benefit from ICD therapy
Study type: Meta- than those with more preserved
analysis of RCTs left ventricular function.

Size: 3 RCTs
MAVERIC Aim: to test the Inclusion criteria: Intervention: EP- 1 endpoint: Of the 108 61% of patients had prior MI
Lau et al. 2004 possibility of survivors of sustained guided interventions EP arm patients, 31 (29%) EPS has a minimal impact on the
(135) prospectively VT, VF or SCD in the (AAD, coronary received an ICD, 46 (43%) diagnosis of patients presented
15172648 identifying patients absence of an AMI in revascularization, received AAD only (mainly with VT, VF or SCD.
who would benefit the last 48h. and ICD) (106 amiodarone or sotalol) The trial does not support a role for
most ICD by EPS in and 18 (17%) received EP testing in risk stratification.

67

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
the context of 2 Exclusion criteria: life patients assigned to coronary
prevention. expectancy of <6 mo this arm) revascularization but no
from a non- ICD. No significant
Study type: RCT arrhythmic cause or Comparator: differences in survival or
child-bearing age therapy with arrhythmia recurrence
Size: 214 patients amiodarone (108 existed between the two
patients assigned to treatment arms after 6 y.
this arm) However, ICD recipients
had a lower mortality
than non-ICD recipients,
regardless of allocated
treatment (HR=0.54,
p=0.0391).
Claro et al. Aim: To evaluate Inclusion criteria: Intervention: 1 endpoint: For 2 Conclusions: With very low
2015 (136) the effectiveness Randomized assessing Amiodarone prevention, amiodarone quality evidence, amiodarone leads
26646017 of amiodarone for the efficacy of compared to placebo or to a statistically non-significant
1 or 2 prevention amiodarone vs. Comparator: no intervention (two increase in the risk of SCD and all-
of SCD compared placebo, no placebo, no studies, 440 participants) cause mortality (by 33% to 600%)
with placebo or no intervention, or other intervention, ICD or appeared to increase the when compared to placebo or no
intervention or any antiarrhythmics in other risk of SCD (RR: 4.32; 95% intervention. This meta-analysis
other adults, either for 1 antiarrhythmics CI: 0.8721.49) and all- did not effectively rule out benefit
antiarrhythmic. prevention or 2 cause mortality (RR: or harm for 2 prevention with
prevention of SCD. 3.05;95% CI: 1.337.01). amiodarone.
Study type: meta- Compared to other AAD Side effects: Amiodarone was
analyses using a (four studies, 839 associated with an increase in
random-effects participants) amiodarone pulmonary and thyroid adverse
model appeared to increase the events.
risk of SCD (RR: 1.40; 95% Limitations: For 2 prevention,
Size: 24 studies CI: 0.563.52; very low the evidence is inconsistent and
(9,997 participants) quality of evidence), but the quality of the evidence was
with 6 studies there was no effect in all- very low, so the authors concluded
identified as 2 cause mortality (RR: 1.03; that there is uncertainty on the
prevention trials. 95% CI: 0.751.42; low findings. There are some
quality evidence). methodological issues that warrant
certain caution when interpreting
these results.

68

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 18. Nonrandomized Trials, Observational Studies, and/or Registries for Secondary Prevention Sudden Death in Ischemic
Heart Disease (Section 7.1.1)
Study Acronym; Aim of Study; Patient Population Endpoint and Results Relevant 2 Endpoint (if any);
Author; Study Type; Study Limitations;
Year Published Study Size (N) Adverse Events
Raitt et al. 2001 Aim: To determine Inclusion criteria: 1 endpoint: Mortality Sustained VT without serious
(137) prognostic implications of Patients with stable VT symptoms or hemodynamic
11208684 stable VT that were not enrolled in Results: The mortality in 440 compromise is associated with a
AVID, were included in a patients with stable VT tended to high mortality rate and may be a
Study type: Observational, registry of patients be greater than that observed in marker for a substrate capable of
registry of patients with screened for the study. 1029 patients presenting with producing a more malignant
hemodynamically stable unstable VT (33.6% vs. 27.6% at 3 arrhythmia.
VT Exclusion criteria: y; RR:1.22; p=0.07). After
Patients who had an adjustment for baseline and
Size: The study population arrhythmia within 5 d of a treatment differences, the RR was
consisted of 440 patients MI, cardiac surgery, or little changed (RR: 1.25, p=0.06).
with stable VT and 1029 coronary intervention
patients with unstable VT. were excluded, as were
Of the 1029 patients with patients with class IV HF
unstable VT, 330 had or those who were on a
therapy determined by heart transplant list, had
randomization in the AVID a prior ICD implant or
trial: 52% received an ICD, attempted implant, or
47% amiodarone, and 2% had a life expectancy of
sotalol. Therapy for the <1 y.
remaining 699 patients
with unstable VT and the
440 patients with stable VT
was determined at the
discretion of the attending
physician.

69

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Bass EB et al. Study type: retrospective Inclusion: unexplained Results: Conclusion: patients with
1988 (138) cohort syncope EP study EP study had positive results in 37 electrophysiologically positive
3195480 between April 1981 and patients--31 with VT, 3 with SVT results had high rates of SCD and
Size: 70 patients April 1986. and 3 with abnormal conduction. total mortality
Exclusion: N/A
No difference in the 3 y recurrence
rate between the studies (32 vs
24%, respectively).

At 3 y, patients + had higher rates


of SCD than patients with - results
(48% vs 9%, respectively,
p<0.002).

3 y total mortality rate was also


higher with + results than among
those with - (61% vs 15%,
respectively, p<0.001).
Owens DK et al. Aim: Evaluated whether Markov model to Results: cost-effectiveness The cost-effectiveness of ICD use
2002 (139) risk stratification based on evaluate the cost- becomes unfavorable at both low relative to amiodarone depends on
12228780 risk ofSCD alone was effectiveness of ICD and high total cardiac mortality total cardiac mortality rates as well
sufficient to predict the implantation compared rates. as the ratio of sudden to
effectiveness and cost- with empiric amiodarone If the annual total cardiac nonsudden cardiac death.
effectiveness of the ICD. treatment. The model mortality rate is 12%, the cost-
incorporated mortality effectiveness of the ICD varies
rates from sudden and from $36,000 per quality-adjusted
nonsudden cardiac death, life-year (QALY) gained when the
noncardiac death and ratio of sudden cardiac death to
costs for each treatment nonsudden cardiac death is 4 to
strategy. Model assumed $116,000 per QALY gained when
that the ICD reduced total the ratio is 0.25.
mortality rates by 25%,
relative to use of
amiodarone.

70

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 19. Nonrandomized Trials, Observational Studies, and/or Registries for Coronary Artery Spasm (Section 7.1.1.1)
Author; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Year Published Study Size (P values; OR or RR; Comment(s)
& 95% CI)
Ahn et al. 2016 Study type: 188 patients with variant 1 endpoint: The 1 end point cardiac Conclusions: The prognosis of
(140) retrospective angina with aborted SCD death patients with variant angina with
27386766 multicenter cohort and 1,844 patients with ASCD was worse than other
variant angina without Cardiac death was significantly higher patients with variant angina. In
Size: 188 patients with aborted SCD from 13 heart in aborted SCD patients (24.1 /1,000 addition, our findings supported
aborted SCD centers in South Korea. patient-y vs. 2.7/ 1,000 patient-y (HR: ICDs in these high-risk patients as a
7.26; 95% CI: 4.21-12.5; p<0.001) 2 prevention because current
Median followup of 7.5 multiple vasodilator therapy
y Predictors included family Hx of SCD appeared to be less optimal.
(OR: 3.67; 95% CI: 1.27-10.6; p=0.016), Limitations: Retrospective study
multivessel spasm (OR: 2.06; 95% CI: and no accurate information for
1.33-3.19; p=0.001), and LAD artery response to medical therapy or
spasm (OR: 1.40; 95% CI: 1.02-1.92; compliance. This is an ethnically
p=0.04) homogenous group raising
questions about extrapolation to
A total of 24 aborted SCD patients other ethnicities. It is unknown
received ICD what factors might have led
physicians to implant an ICD.
6 ICD patients experienced VF and 1
died due to intractable VF.

In the aborted SCD patients who


received an ICD, mortality was 4.3%
compared with 19.3% of those that did
not receive an ICD (trend but
nonsignificant p=0.15)
Yamashina et Study type: Resuscitated from CA with 1 endpoint: recurrent VT/VF Conclusions: Medical therapy
al. 2014 (141) retrospective single 1) documented VF/VT or associated with favorable long-
23906527 center cohort PEA and 2) the absence of Results: No recurrent VA, syncope, or term outcomes for patients with
significant narrowing due CA during a mean followup of 67 mo vasospastic angina associated with
Size: 18 patients in to coronary (1 of 18 died during the initial CA.
Japan between 1992 atherosclerosis or any hospitalization and another cancer).
and 2012 structural cardiac All are treated with long-acting

71

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
abnormalities possibly CCBs/nitrates and successfully quit Limitations: small, retrospective,
causing CA; 3) absence of smoking. and non-randomized study in a
identifiable or reversible single Japanese center.
causes of lethal VA 4) 6 received ICD none received
documented ST elevation therapies
during chest pain or
positive provocation test
Eschalier et al. Study type: case Patients with CA related to Results: 2/3 patients underwent ICD Conclusions: Very small case series
2014 (142) reports coronary artery vasospasm implantation because of recurrent VT demonstrating ICD use in patients
24373622 despite medical therapy. None had with coronary vasospasm.
Size: 3 patients. ICD shocks in follow-up.
Matsue et al. Study type: 23 patients with aborted Endpoints: Appropriate ICD therapy, Results: The average time for
2012 (143) retrospective SCD receiving a 2 sudden CA, or death from all causes appropriate ICD therapy from ICD
22840527 observational cohort prevention ICD in the insertion was about 1 y and only
absence of SHD or CAD 26% of patients experienced event 2/5 patients with recurrent lethal
Size: 23 patients. from who had spasm of a major arrhythmia had symptoms of chest
3 Japanese hospitals epicardial coronary artery 4 patients had an episode of VF pain prior to ICD therapy.
induced with acetylcholine appropriately treated by their ICD and Conclusions: These data support
Mean followup period challenge survived (all but 1 patient was the use of ICD therapy in patients
of 2.9 y compliant with vasodilator therapy). with coronary artery vasospasm
After the first episode of appropriate who have survived an episode of
ICD therapy in these 4 patients, none life-threatening VT/VF
received recurrent therapy during the Limitations: Non-randomized
limited follow-up. and relatively small number of
Japanese patients in only 3
1 additional patient survived CA 2 to cardiovascular centers.
pulseless electrical activity The cohort in the present study
included only patients with
coronary vasospasm who had SCD,
and thus the data shown here
cannot be extrapolated to the
whole coronary vasospasm
population.
Medication compliance was
evaluated only by medical
interview with patients, and that

72

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
may have caused over-estimation
of compliance.
Takagi et al. Study type: nationwide 30 men and 5 women had 1 endpoint: The 1 end point MACE Results (continued): In the 35
2011 (144) registry of patients with OHCA within a registry of included cardiac death, nonfatal MI, OHCA survivors, 14 patients
21406685 vasospastic angina 1429 patients in Japan hospitalization for unstable angina underwent ICD implantation while
with vasospastic angina pectoris and HF, and appropriate ICD intensively treated with calcium
Size: 35 patients with (definition: an angina shocks during the follow-up period, channel blockers.
OHCA. attack at rest and/or on which began at the date of original Appropriate ICD shocks for VF in 2
effort, accompanied by a VSA diagnosis. of 14 patients despite intensive
transient ECG ST-segment medical treatment. SCD occurred
elevation or depression of 2 endpoint: The 2 end point was all- in 1 patient without an ICD who
>0.1mV or a newly cause mortality. self-discontinued medication prior
appearance of negative U to the fatal event.
wave in at least 2 related Results: Rate of cardiac death and
leads, and/or a total or Survival rate free from MACE was nonfatal MI in patients in whom
subtotal coronary artery significantly lower in the OHCA medications were reduced or
narrowing during the survivors compared with the non- discontinued (8%, 2 of 25 patients)
provocation test of OHCA patients (72% vs. 92% at 5 y, was 10-fold higher than that in the
coronary spasm, p<0.001). There was no difference in patients with continued
accompanied by chest all-cause mortality between the medications (0.7%, 10 of 1404
pain and/or ischemic ECG groups. patients, p=0.017).
changes mentioned Limitations: Appropriate ICD
above) therapy is used as surrogate for
sudden death. Retrospective
observational study and there the
association found in the present
study is not necessarily causal and
follow-up duration was variable
possible many arrhythmic events
were missed.
Meisel et al. Study type: Inclusion criteria: (1) Results: All patients were treated with Conclusions: VF complicating
2002 (145) Retrospective case typical chest pain at rest maximum tolerated calcium channel variant angina is a higher risk
11988204 review with multicenter associated with transient antagonists. population. Raises possibility that
survey ST-segment elevations not some patients such as those
present on the baseline Ventricular arrhythmia reoccurred remaining symptomatic despite
ECG and disappearing with after discharge in all patients. Median medical therapy should be
relief of pain; (2) time to the first arrhythmia recurrence considered for an ICD.

73

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 8 patients with documented VF was 15 mo (range 2-112). An ICD was
vasospastic angina immediately after the subsequently implanted in 7 patients.
complicated by VF ischemic episode; (3)
survival of the index After ICD implantation, 4 patients
episode of VF; (4) received appropriate ICD shocks for
angiographically normal VT/VF. 1 patient died with ICD and
coronary arteries defined recurrent chest pain with EMD.
as patent arteries with no
irregularities; (5) 1 patient with recurrent VF and no ICD
angiographic evidence of had recurrent VF out of hospital and
coronary spasm defined as subsequent brain damage and died
transient narrowing of several years later.
arterial lumen or recurrent
episodes of ECG
documented ischemia
especially if occurring in
different coronary
territories; and (6)
recurrent angina despite
medical therapy

Exclusion criteria: N/A


Chevalier et al. Study type: Inclusion criteria: Results: At a mean follow-up 58 mo, 6 Conclusions: medical treatment
1998 (146) retrospective case survivors of CA with patients remained free of symptoms. 1 with calcium channel antagonists
9426018 review positive ergonovine patient who continued smoking had a appears to be associated with an
provocation test new CA despite 10 y after and was event-free clinical course. Stopping
Size: 7 patients discovered to have a new LAD and RCA smoking is important.
Mean age was 44 y; 3 stenosis and underwent CABG and ICD
were male and 4 females. placement.
All of them were habitual
cigarette smokers.

Exclusion criteria: N/A


Myerburg et al. Study type: Inclusion: From 356 Results: Titration of calcium channel Conclusions:
1992 (147) retrospective cohort patients, included were 5 blocking drugs (verapamil, diltiazem, Silent MI due to coronary artery
1574091 survivors of OHCA or nifedipine) against the ability of spasm can initiate potentially fatal
Size: 5 patients between 1980 and 1991 ergonovine to provoke spasm was

74

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
without epicardial CAD successful in preventing recurrent arrhythmias in patients without
with induced or arrhythmias in all 4 patients. flow-limiting CAD.
spontaneous focal In patients with OHCA due to
coronary artery spasm (or 1/5 patients had a positive EPS with coronary vasospasm, treatment
both) ventricular flutter despite propranolol with calcium channel blocking
Exclusion criteria: N/A so ICD was implanted. agents appears to prevent
recurrent arrhythmias.

Data Supplement 20. Nonrandomized Trials, Observational Studies, and/or Registries for Post CABG VT/VF (Section 7.1.1.2)
Author; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Year Published Study Size (P values; OR or RR; Comment(s)
& 95% CI)
Saxon et al. Study type: 17 patients UCLA VT/VF patients had lower LVEF, more Conclusions:
1995 (148) retrospective single medical center with likely to have had MI <2 w before New onset MMVT is usually associated
7856540 center cohort new-onset sustatined CABG, graft to chronically occluded with old infarct/scarring (and many
VT/VF within 30 d of vessel inducible at EPS)
Size: 17 patients CABG between 1981- Polymorphic VT/VF usually associated
1993 compared to Sustained MMVT 11/17 patients with ischemia.
119 control patients (65%) and most (64%) had no Polymorphic VT/VF occurring after
1992-1993 without evidence of peri-op MI. Those with CABG warrants a therapeutic approach
VT/VF post-CABG MMVT, 80% inducible at EPS targeting treatment of MI.

Polymorphic VT/VF 6/17 patients


(35%) and most had peri-op MI (67%)
and only 2/6 (33%) had inducible VT
at EPS
Ascione et al. Study type: Cases CABG patients Factors associated with VT/VF age Results (cont.):
2004 (149) retrospective single 4/1996-9/2001 with <65 y, female, low BMI, unstable 5/12 (42%) intraoperative VT/VF died in
15120824 center cohort VT/VF post-op angina, reduced LVEF, and need for the hospital, as compared with 10/55
compared to controls inotrope or IABP (18%) with VT/VF in post-op period
Size: 4411 patients without. Assessed (p=0.08). Those with post-op VT/VF, 27
undergoing CABG

75

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
including 69 patients factors associated Off-pump CABG associated with (47.4%) had the event within the first 24
with post op VF/VT with post-op VT/VF protective effect (OR: 0.53; 95% CI: h.
0.251.13) Conclusion: incidence of VT/VF is low in
None of the VT/VF patients undergoing CABG but associated
patients underwent Long term survival was similar with high in-hospital mortality. The late
ICD placement. between groups (2 y 98.2% VT/VF survival of those discharged is similar to
surviving to discharge vs. 97% for controls.
control (HR: 0.96; 95% CI: 0.42.3)
Steinberg et al. Study type: cohort Patient with sustained Results: 12 patients (3.1%) Results (cont.): Patients with VT were
1999 (150) study post-op VT 24 hrs experienced 1 episode of sustained more likely to have prior MI (92% vs.
10027813 but <30 d after CABG VT 4.14.8 d after CABG 50%, p<0.01), severe CHF (56% vs. 21%,
Size: 12 patients among consecutive p<0.01), and LVEF <0.40 (70% vs. 29%,
patients 382 patients In 11 /12 patients, no postoperative p<0.01).
undergoing CABG at a complication explained the VT. 1 By multivariate analysis, the number of
single institution patient had a perioperative MI. bypass grafts across a noncollateralized
occluded vessel to an infarct zone was
Variables associated The in-hospital mortality rate was the only independent factor predicting
with the occurrence 25%. Among the 9 survivors, 5 had VT.
of VT was performed EPS with all inducible sustained Conclusions: (1) Patients who
monomorphic VT (matching clinical developed VT had a high in-hospital
VT). 3/9 patients received an ICD mortality rate of 25% (2) However, long-
before hospital discharge. Other 6/9 term outcome was good (possibly related
patients received chronic therapy to antiarrhythmic or ICD). (3) predictors
with AAD (primarily amiodarone). are MMVT previous MI scar and
associated severe LV dysfunction. (4)
All 9 patients are alive, with a mean Relationship was found between the
followup of 2.5 y. development of VT and the placement of
a bypass graft across a noncollateralized
2 patients (1 with an ICD and 1 on occluded coronary vessel to a chronic
amiodarone) had recurrent VT during infarct zone. (5) The development of
follow-up. MMVT was typically not due to a
detectable postoperative complication or
ischemia.

76

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 21. RCTs and Nonrandomized Trials, Observational Studies, and/or Registries of ICDs Primary Prevention Ventricular
Arrhythmias and Sudden Death in Patients with Ischemic Cardiomyopathy (Section 7.1.2)
Study Aim of Study; Patient Population Randomized Subjects Endpoint and Conclusion:
Acronym; Study Type; Results
Author; Study Size (N)
Year Published
MADIT-I Aim: To evaluate Inclusion: Previous MI, LVEF 35%, Comparator: All-cause mortality: In patients with a prior
Moss et whether NSVT, inducible VT at EPS that was Control (101 patients) Control 32% vs. ICD MI, low EF who are at
al.1996 (42) prophylactic ICD, non-suppressed with IV procainamide 13% high risk for VT,
8960472 as compared with or equivalent AAD Intervention: (RRR -59% ARR - prophylactic therapy
conventional ICD (95 patients) 19%) with an ICD leads to
medical therapy, Exclusion: previous CA or VT causing improved survival as
would improve syncope that was not associated with compared with
survival in a high- an AMI; symptomatic hypotension conventional medical
risk group of while in a stable rhythm; and MI <3 therapy.
patients with wk, prior CABG <2 mo or PCI <3 mo,
NSVT, reduced as were women of childbearing age
LVEF and previous who were not using medically
MI. prescribed contraceptives, patients
with advanced cerebrovascular
Study type: disease, patients with any condition
prospective other than cardiac disease that was
multicenter RCT associated with a reduced likelihood
of survival for the duration of the trial,
Size: 196 patients and patients who were participating
in other clinical trials
CABG-Patch Aim: To evaluate Inclusion: Coronary artery bypass Comparator: All-cause mortality: No evidence of
Bigger et the role of ICD in surgery, EF <36, SAECG positive Control (454 patients) Control 18% vs. ICD improved survival among
al.1997 (151) patients after 18% patients with CAD,
9371853 CABG with high Exclusion: sustained VT/VF, diabetes Intervention: reduced LVEF, and
risk of SCD mellitus with poor blood glucose ICD (446 patients) abnormal SAECG
control or recurrent infections, receiving prophylactic
Study type: RCT previous or concomitant aortic- or ICD after CABG
mitral-valve surgery, concomitant
Size: 900 patients cerebrovascular surgery, a serum
creatinine concentration greater than
3 mg/dl, emergency CABG, a

77

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
noncardiovascular condition with
expected survival of less than 2 y, or
an inability to attend followup visits

MUSTT Aim: To evaluate Inclusion: CAD, LVEF 40%, NSVT, If sustained VT/VF Risk of CA or death Patients with CAD, left
Buxton et al. the usefulness of inducible at EPS were induced by EPS, from arrhythmia ventricular dysfunction,
2000 (41) EPS for risk patients were among the patients and asymptomatic, NSVT
10874061 stratification Exclusion: H/o of syncope or had randomized to who received in whom sustained VAs
among patients sustained VT/VF >48 h after the onset antiarrhythmic treatment with ICDs cannot be induced have
with CAD, of AMI, NSVT that occurred only in therapy, including was lower than that a significantly lower risk
abnormal the setting of drug-induced LQTS or AAD and possible ICD, among the patients of SCD and lower overall
ventricular AMI or that was attributable to acute as indicated by the discharged without mortality than similar
function, and metabolic disorders or drug toxicity, results of EP testing, (HR: 0.24; 95% CI: patients with inducible
NSVT or if they had symptomatic NSVT or no antiarrhythmic 0.130.45; p<0.001) sustained
therapy. All-cause mortality: tachyarrhythmias.
Study type: RCT Control 55% vs. ICD Important to point out
Comparator: 24% that receipt of an ICD
Size: 704 patients Control (353 patients) (RRR -58% and ARR - was not randomized
Inducible but no 31%) treatment.
antiarrhythmic

Intervention:
Inducible and failed
suppression with AAD
and given ICD (161
patients)
MADIT-II Aim: To evaluate Inclusion: Prior MI (>1 mo), EF 30% Comparator: All-cause mortality: In patients with a prior
Moss et al. the benefit of ICD Control (490 patients) control 22% vs. ICD MI and advanced left
2002 (44) in patients with Exclusion: existing indication for ICD; 16% ventricular dysfunction,
11907286 prior MI and NYHA class IV at enrollment; had Intervention: (RRR -28% and ARR - prophylactic ICD
reduced LVEF undergone coronary revascularization ICD (742 patients) 6%) improves survival and
<3 mo; MI <30 d, advanced should be considered as
Study type: RCT cerebrovascular disease, childbearing a recommended therapy.
age and not using contraceptive,
Size: 1232 patients presence of any condition other than
cardiac disease that was associated
with a high likelihood of death during
78

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
the trial, or unwilling to provide
consent

DINAMIT Aim: Inclusion: Recent MI (6-40 d), EF Comparator: All-cause mortality: Prophylactic ICD
Hohnloser et To assess the 35%, standard deviation of normal- Control (342 patients) control 17% vs. ICD therapy does not reduce
al. 2004 (152) benefit of ICD in to-normal RR intervals of 70 msec or 19% overall mortality in high-
15590950 patients with less or a mean RR interval of 750 msec Intervention: risk patients who have
recent MI and or less, mean heart rate 80 ICD (332 patients) recently had a MI.
reduced LVEF beats/min 2 outcome: Although ICD therapy
Study type: RCT arrhythmic death: was associated with a
Exclusion: CHF class IV; noncardiac 12 ICD group vs. 29 reduction in the rate of
Size: 674 patients disease that limited life expectancy; in the control group death due to arrhythmia,
CABG performed since the qualifying (HR ICD group, 0.42; that was offset by an
infarction or planned to be performed 95 95% CI 0.22 to increase in the rate of
within 4 wks after randomization; 0.83; p=0.009) death from
three-vessel PCI performed since the nonarrhythmic causes.
qualifying infarction; name on a
waiting list for a heart transplant;
current, ongoing ICD therapy; prior
implantation of a permanent
pacemaker; requirement for an ICD
(i.e., sustained VT or fibrillation more
than 48 h after the qualifying
infarction); low probability that the
study ICD could be implanted within 7
d after randomization; and expected
poor compliance with the protocol
SCD-HeFT Aim: Evaluate Inclusion: NYHA class I-III HF, LVEF Intervention 1: All-cause mortality: In patients with NYHA
Bardy et al. whether 35% GDMT plus a ICD (829 control 36% vs. ICD class II or III HF and
2005 (43) amiodarone or a patients) 29% LVEF35%, amiodarone
15659722 conservatively Exclusion: Age <18 y, unable to give (RRR: -23% and ARR: has no favorable effect
programmed consent Intervention 2: -7%) on survival, whereas
shock-only, single- GDMT plus single-lead, shock-only
lead ICD would amiodarone (845 ICD therapy reduces
decrease the risk patients) overall mortality. This
of death from any was the longest and
cause in a broad Comparator 1: largest ICD trial.
79

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
population of GDMT plus Placebo
patients with mild- (847 patients)
to-moderate HF

Study type:
prospective
multicenter RCT

Size: 2521 patients


IRIS Aim: Test Inclusion: Recent MI (5-31 d) plus HR Comparator: All-cause mortality: Prophylactic ICD
Steinbeck et whether patients >90 bpm and LVEF 40% or NSVT Control (453 patients) control 23% vs. 22% therapy did not reduce
al. 2009 (153) at increased risk overall mortality among
19812399 who are treated Exclusion: VAs that occurred before Intervention: patients with AMI and
early with an ICD the index MI or >48 h after the MI and ICD (445 patients) clinical features that
will live longer that required treatment, NYHA class placed them at increased
than those who IV drug-refractory HF, an interval of risk.
receive GDMT >31 d between MI and presentation,
alone no ECG documentation within <48 h
after the onset of chest pain, an
Study type: indication for CABG before study
prospective RCT entry, a psychiatric disorder, severe
concomitant disease, a Hx of poor
Size: 898 patients compliance with treatment, either the
inability to participate in this trial or
current participation in another trial,
and an unstable clinical condition
Piccini et al. Aim: To evaluate Inclusion criteria: Studies in which 1 endpoint: SCD, Amiodarone Conclusions:
2009 (154) the cumulative patients were randomized to CVD, all-cause reduces the risk of Amiodarone reduced the
19336434 evidence amiodarone and placebo or inactive mortality, and the SCD by 29% and risk of SCD but is neutral
regarding the control. Additional incidences of drug CVD by 18%, with respect to all-cause
safety and efficacy inclusion criteria included: treatment toxicities. however, mortality.
of amiodarone in for >30 d, followup >6 mo, and amiodarone therapy
prevention of SCD availability of all-cause mortality as an Results: Amiodarone is neutral with Authors suggested
endpoint decreased the respect to all-cause amiodarone as a viable
Study type: Meta- incidence of SCD mortality alternative in patients
analysis of all RCT Exclusion criteria: Studies (7.1% vs. 9.7% [OR: who are not eligible for
examining the use 0.71; 95% CI: 0.61 or who do not have

80

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
of amiodarone vs. of patients with shock-refractory VA, 0.84, p<0.001]) and Adverse events: access to ICD therapy for
placebo/control OHCA, patients <18 y, randomization cardiovascular death associated with a 2- the prevention of SCD.
for the prevention to amiodarone vs. a class Ic or class III (14.0% vs.16.3% [OR: and 5-fold increased
of SCD AAD (without a placebo or standard of 0.82;0.710.94, risk of pulmonary
care arm). Studies of patients with p=0.004]). There was and thyroid toxicity.
Size: 15 trials, ICDs were excluded unless used on a 1.5% absolute risk
which randomized both arms. reduction in all-cause
8,522 patients mortality which did
not meet statistical
significance (p=0.093).
Amiodarone therapy
increased the risk of
pulmonary (2.9% vs.
1.5% [OR: 1.97;95%
CI:1.273.04,
p=0.002]), and thyroid
(3.6% vs. 0.4%; [OR:
5.68; 95% CI :2.94
10.98, p<0.001])
toxicity.
Claro et al. Aim: To evaluate Inclusion criteria: Randomized Intervention: 1 endpoint: There Conclusions: There is
2015 (136) the effectiveness assessing the efficacy of amiodarone Amiodarone was a beneficial low quality evidence that
26646017 of amiodarone for vs. placebo, no intervention, or other effect with amiodarone reduces the
1 or 2 antiarrhythmics in adults, either for 1 Comparator: placebo, amiodarone risk of SCD and may
prevention of SCD prevention or 2 prevention of SCD. no intervention, ICD reducing the risk of reduce the risk of all-
compared with or other SCD by 12%-34% cause mortality when
placebo or no antiarrhythmics and may reduce the compared with placebo
intervention or risk of all-cause or no intervention in a 1
any other mortality by up to prevention setting.
antiarrhythmic. 22% when
compared with The evidence regarding
Study type: placebo or no the comparison with
meta-analyses intervention in a 1 other antiarrhythmics is
using a random- prevention setting. of moderate quality and
effects model goes in the same
direction.

81

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 24 studies Adverse events: Stresses the
(9,997 Amiodarone was importance for people in
participants) with associated with low-income countries,
17 studies with increased adverse where an ICD may not be
8383 patients effects, both thyroid available.
identified as and pulmonary
relevant 1 (based on 12
prevention trials. studies), and
increased risk of
discontinuation
(based on 13
studies) when
compared with
placebo.
Owens DK et Aim: Evaluated Markov model to evaluate the cost- Results: cost-effectiveness becomes The cost-effectiveness
al. 2002 (139) whether risk effectiveness of ICD implantation unfavorable at both low and high total of ICD use relative to
12228780 stratification compared with empiric amiodarone cardiac mortality rates. amiodarone depends on
based on risk treatment. The model incorporated If the annual total cardiac mortality rate is total cardiac mortality
ofSCD alone was mortality rates from sudden and 12%, the cost-effectiveness of the ICD varies rates as well as the ratio
sufficient to nonsudden cardiac death, noncardiac from $36,000 per quality-adjusted life-year of sudden to nonsudden
predict the death and costs for each treatment (QALY) gained when the ratio of sudden cardiac death.
effectiveness and strategy. Model assumed that the ICD cardiac death to nonsudden cardiac death is
cost-effectiveness reduced total mortality rates by 25%, 4 to $116,000 per QALY gained when the
of the ICD. relative to use of amiodarone. ratio is 0.25.
Cantero- Aim: To evaluate Inclusion criteria: Results: Appropriate ICD
Prez EM, et al. the effectiveness Records from patients accepted for Median follow-up of 77 d therapies were recorded
2013 (155) of ICDs for primary heart transplantation from January 1, overall mortality in the ICD group was 7.1% in 42.9% (12/28) in this
24314988 prevention in 2006, to July 30, 2012, and whose (2/28) and in the non-ICD group was 17.6% population.
patients with LVEF LVEF was <31% were reviewed (9/51; p=0.062).
30% included on Cause of death in patients without ICDs:
the heart Sudden death (5/9, 55.6%),
transplantation list HF (4/9, 44.4%).
Cause of death in patients with ICDs: HFheart
Size: Patients who
received ICDs for
primary
prevention (N=28)

82

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
were compared
with patients
without ICDs
(N=51)
Frhlich GM, Aim: To delineate Inclusion criteria: Results: ICDs appear to be
et al. 2013 the role of ICD Patients listed for heart Median time on the waiting list = 8 mo associated with a
(156) therapy for the transplantation in 2 tertiary heart (estimated 1-year: 883% vs. 773% vs. reduction in all-cause
23813845 primary and transplant centres were enrolled. Of 673%; p=0.0001). mortality in patients
secondary 550 patients (51%) on the transplant An independent beneficial effect of ICDs that implanted with the
prevention of SCD list with an ICD: was most pronounced in patients who had device for primary and
in patients listed primary prevention ICD: N=216 received an ICD for primary prevention (HR: secondary prevention
for heart secondary prevention ICD: N=334 0.4, 95% CI: 0.190.85; p=0.016). compared to those
transplantation without an ICD.

Size: N=1089
Gandjbakhch Aim: To evaluate Inclusion criteria: Results: Need for mechanical
E, et al. 2016 the ICD benefit on Patients with end-stage HF receiving 15.6% of patients died while awaiting heart circulatory support
(157) mortality in an ICD before or within 3 mo after transplantation. (p<0.001), low EF
27344378 patients with end- being listed for heart transplantation Non-ICD patients presented more often (p=0.001) and
stage HF listed for haemodynamic compromise. registration on the
heart ICD did not remain an independent predictor regular list (p=0.008)
transplantation of death. were the only
Death by haemodynamic compromise (76.3% independent predictors
Size: N=380 of deaths), which occurred more frequently of death.
consecutive in the non-ICD group (14.7% vs. 5.8%; log- ICD-related
patients listed for rank p=0.002). complications occurred
heart in 21.4% of patients,
transplantation Unknown/arrhythmic deaths did not differ mainly as a result of
between 2005 and significantly between the two groups (3.9% postoperative worsening
2009 in A tertiary vs. 1.7%; log-rank p=0.21). of HF (11.9%).
heart transplant
centre
Vakil K, et al. Aim: To assess the Inclusion criteria: Results: In the subgroup of
2016 (158) impact of ICD on Adults (age 18 y) listed for first-time Median follow-up of 154 days, patients with LVAD (N=
waitlist mortality heart transplantation in the US 3,638 patients (11%) died on the waitlist (9% 9,478), having an ICD was
in patients listed between January 1, 1999, and in ICD group vs. 15% in no-ICD group; associated with an
September 30, 2014, were adjusted 19% relative

83

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
for heart retrospectively identified from the p<0.0001), whereas 63% underwent heart reduction in mortality
transplantation United Network for Organ Sharing transplantation. (HR: 0.81; 95% CI: 0.70
registry. An ICD at listing was associated with an 0.94).
Size: N=32,599 adjusted 13% relative reduction in mortality
(HR: 0.87; 95% CI: 0.800.94).

Data Supplement 22. RCTs Evaluating Treatment and Prevention of Recurrent Ventricular Arrhythmias in Patients with Ischemic Heart
Disease (Section 7.1.3)
Study Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if any);
Acronym; Study Type; (# patients) / (Absolute Event Study Limitations;
Author; Study Size (N) Study Comparator Rates, Adverse Events
Year Published (# patients) P values; OR or RR; &
95% CI)
OPTIC Aim: Determine Inclusion criteria: Patients Intervention: 1 endpoint: ICD Amiodarone plus BB
Connolly et whether who had received an ICD amiodarone plus BB shock for any reason. significantly reduced the risk of
al. 2006 (159) amiodarone plus BB within 21 d for inducible or or sotalol Shocks occurred in 41 shock compared with BB alone
16403928 or sotalol are better spontaneous VT/VF patients (38.5%) (HR: 0.27; 95% CI: 0.140.52;
than BB alone for Comparator: BB alone assigned to BB alone, p<0.001) and sotalol (HR: 0.43;
prevention of ICD Exclusion criteria: Long QT 26 (24.3%) assigned 95% CI: 0.220.85; p=0.02).
shocks. syndrome, corrected QT to sotalol, and 12 There was a trend for sotalol to
interval of more than 450 (10.3%) assigned to reduce shocks compared with
Study type: RCT ms, already receiving or amiodarone plus BB BB alone (HR: 0.61; 95% CI:
recent treatment with a (HR: 0.44; 95% CI: 0.371.01; p=0.055).
Size: 412 patients class I or class III 0.280.68; p<0.001). Adverse pulmonary and
antiarrhythmic agent, thyroid events and
creatinine clearance less Safety endpoint: NA symptomatic bradycardia were
than 30 mL/min, AF likely to more common among patients
require use of a class I or randomized to amiodarone.
class III antiarrhythmic
agent, absence of SHD, Conclusions: Despite use of
NYHA class IV HF advanced ICD technology and
treatment with a BB, shocks
occur commonly in the first
year after ICD implant.
Amiodarone plus BB is effective
84

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
for preventing these shocks
and is more effective than
sotalol but has an increased
risk of drug-related adverse
effects.
Pacifico et al. Aim: Efficacy and Inclusion criteria: age >18 y, Intervention: 160 to 1 endpoint: First inappropriate shock for
1999 (160) safety of sotalol to life-threatening VT that were 320 mg of sotalol per Treatment with a SVT or death from any cause
10369848 prevent shocks from not due to a reversible day sotalol was was reduced with sotalol
ICDs cause; had received their associated with a (reduction in risk, 64%;
first or a replacement ICD Comparator: lower risk of death p=0.004).
Study type: within 3 mo before matching placebo from any cause or the Sotalol also reduced the
prospective, RCT enrollment (patients with delivery of a first mean frequency of shocks due
double-blind replacement defibrillators shock for any reason to any cause (1.433.53
had to have received at least (reduction in risk shocks/y, as compared with
Size: 302 patients one shock during the 48%; p<0.001; first 3.8910.65 in the placebo
preceding 6 mo); had a ICD appropriate shock for group; p=0.008).
that provided tiered therapy a va or death from
with EGM and separate any cause was also Conclusions: Oral sotalol was
logging of shocks reduced (reduction in safe and efficacious in reducing
risk, 44%; p=0.007), the risk of death or the delivery
Exclusion criteria: incessant of a first defibrillator shock
VT; had received AAD Safety endpoint: whether or not ventricular
therapy <5 half-lives of the Bradycardia was function was depressed.
drug before randomization more common in
in the case of class I and III sotalol group, but
agents (and <3 mo before only 2 patients
randomization in the case of discontinued therapy
amiodarone); had a QT because of it; 3
interval of more than 450 patients in each
msec (or a JT interval of group had HF.
more than 360 msec) in the
absence of drug therapy;
had a LQTS, including
prolongation of the QT
interval in response to
specific drugs; had unstable
coronary syndromes or had

85

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
had an AMI less than two
weeks before screening; had
intractable HF (NYHA class
IV); were candidates for
heart transplantation; or had
a medical condition that was
likely to be fatal in less than
2 y.
Kettering et Aim: Efficacy of Inclusion criteria: ICD Intervention: 40-480 1 endpoint: VT/VF Conclusions: No significant
al. 2002 (161) metoprolol vs. implanted for sustained VT mg sotalol daily recurrence requiring difference in freedom from ICD
12494613 sotalol in or VF ICD intervention; 33 therapies between metoprolol
preventing Comparator: 25-200 events in patients and sotalol group (p=0.68)
recurrent VT in Exclusion criteria: mg daily metoprolol treated with
patients with ICDs Contraindications for tartrate metoprolol vs. 30 in
metoprolol or sotalol; AMI patients receiving
Study type: within the sotalol (p=0.68)
prospective, RCT last 4 wk; unstable angina;
severe concomitant diseases Adverse Events: 5
Size: 100 patients metoprolol and 6
sotalol patients
required dose
reduction for fatigue,
dizziness, HF
Echt et al. Aim: Examine the Inclusion: 6 d - 2 y after MI if Intervention: 1 endpoint: Conclusions: Excess of deaths
1991 (162) mortality and they had an average of 6 encainide or arrhythmic death or due to arrhythmia and deaths
1900101 morbidity after PVCs/h on ambulatory flecainide cardiac arrest due to shock after acute
randomization to electrocardiographic recurrent MI in patients
encainide or monitoring of at least 18 h Comparator: placebo After a mean treated with encainide or
flecainide or their duration, and no runs of VT followup of 10 mo, 89 flecainide. Nonlethal events,
respective placebo. of 15 beats at a rate of patients had died: 59 however, were equally
120 beats/mim. EF 0.55 if of arrhythmia (43 distributed between the active-
Study type: RCT recruited within 90 d of the receiving drug vs. 16 drug and placebo groups.
MI, or EF 0.40s if recruited receiving placebo;
Size: 1498 patients 90 d or more after the MI. p=0.0004)

Exclusion: as above

86

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Seidl et Aim: efficacy of d,l- Inclusion criteria: Patients Intervention: 1 endpoint: Conclusions: The recurrence
al.1998 (163) sotalol and with ICD and Hx of VT/VF metoprolol (mean Actuarial rates for rate of VT in patients treated
9761084 metoprolol in dosage 104+37 mg/d) absence of VT with metoprolol was lower
preventing Exclusion criteria: AMI recurrence at 1 and 2 than in patients treated by d,l-
recurrence of within 1 wk; Comparator: d,l- y were significantly sotalol. No difference in
arrhythmic contraindications sotalol (mean dosage higher in the overall survival
events after ICD for BB; Hx of proarrhythmia 242 109 mg/d) metoprolol group
implantation. caused by d,l-sotalol compared with the
d,l-sotalol group (83%
Study type: and 80% vs 57% and
prospective, RCT 51%, respectively,
p=0.016).
Size: 70 patients
Safety endpoint: HF
led to drug
discontinuation in 9%
in each group.
2 episodes of
proarrhythmia in
sotalol group.
Kuhlkamp et Aim: Evaluate Inclusion criteria: Patients Intervention: Patients 1 endpoint: 25 No difference in total mortality
al. 1999 (164) efficacy of sotalol in with inducible sustained VT whose VT was patients (53.2%) in among the 3 groups
9935007 preventing or VF suppressed on sotalol the ICD-only
recurrences of VT were treated with it; group had a VT/VF Conclusion: Sotalol significantly
Exclusion criteria: non- patients whose VT recurrence in reduces the incidence of
Study type: syncopal sustained VT; was not suppressed comparison to 15 recurrences of sustained VT in
prospective, RCT contraindications to BB; on sotalol received an patients (28.3%) in comparison to no AAD
limited projected survival ICD and were the sotalol group and treatment
Size: 146 patients due to comorbid disease randomized to 15 patients (32.6%) in
treatment with sotalol the
or no antiarrhythmic ICD/sotalol group (p 5
therapy 0.0013).

Comparator: no Safety endpoint:


antiarrhythmic Intolerance to
treatment with
d,lsotalol (overt

87

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
cardiac failure,
symptomatic
hypotension or
Bradycardia)

MADIT-II Study type: Inclusion criteria: ischemic 1 endpoint: The frequency of Conclusion: Beta blockers
substudy Retrospective, cardiomyopathy, EF<30%, Appropriate ICD inappropriate ICD reduce the risk for VT or VF and
Brodine et al. observational randomized to ICD arm therapy for VT/VF; therapy for SVT improve survival in ICD-treated
2005 (165) survival was not significantly patients with ischemic
16125497 Size: 720 patients Exclusion criteria: Patients different among the 3 cardiomyopathy.
who received ICDs who were not randomized to Results: Patients in treatment groups
ICD therapy the top quartile of BB (p=0.32).
doses had a significant
reduction in the risk
of VT or VF requiring
ICD therapy compared
with patients not
receiving BB (HR:
0.48; p=0.02). BB use
was also associated
with significant
improvement in
survival compared
with the nonuse of BB
(HR: 0.4; p<0.01).
SMASH VT Aim: To determine Inclusion criteria: age 18 y Intervention: 1 endpoint Trend towards reduced
Reddy et al. whether with MI at least 1 mo Substrate based After 2 y of follow-up, mortality after 2 y in the
2007 (166) prophylactic previously and a Hx of VF, catheter ablation of ICD therapies ablation group (9% vs 17%,
18160685 substrate based Hemodynamically unstable arrhythmogenic occurred in 12% of p=0.06)
catheter ablation in VT, or Syncope with myocardium during patients randomized
sinus rhythm inducible VT and ICD sinus rhythm (N=64) to catheter ablation No difference in left
decreases ICD implantation and 33% in the ventricular function or NYHA
therapies after MI Comparator: Standard control group (HR functional class during follow-
Exclusion criteria: ICD follow-up (N=64) 0.35; CI 0.150.78, up.
Study type: RCT Treatment with AAD, p=0.007)
prospective ischemia induced VT/VF, or
incessant VT or VF
Size: 128 patients
88

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
VANISH Aim: To determine Inclusion criteria: Prior MI, Intervention: 1 endpoint VT storm occurred in 32
Sapp J. et al. whether catheter ICD implantation, at least 1 Randomized 1:1 to The 1 outcome patients (24.2%) in the ablation
2016 (167) ablation decreases episode of VT during catheter ablation or occurred in 78 of 132 group and 42 patients (33.1%)
27149033 ICD therapies in treatment with amiodarone escalated AAD patients (59.1%) in in the escalated-therapy group
patients with or another class I or class III therapy (escalated- the ablation group (HR: 0.66; 95% CI: 0.420.05
ischemic AAD within the previous 6 therapy group), and in 87 of 127 p=0.08). Appropriate ICD
cardiomyopathy mo (N=132) patients (68.5%) in shocks occurred in 50 patients
with a Hx of VT or Exclusion criteria: Failure to the escalated-therapy (37.9%) and 54 patients
VF despite the use give informed consent Comparator: group. The rate of the (42.5%), respectively (HR: 0.77;
of AAD Escalated drug 1 outcome was 95% CI: 0.531.14; p=0.19).
therapy: Amiodarone significantly lower in
Study type: loading then amio 200 the ablation group 36 patients (27.3%) in the
randomized, mg/d (if on Sotalol) or than in the escalated- ablation group and 35 (27.6%)
prospective Amiodarone reloading therapy group in the escalated-therapy group
then 300 mg/d if on (HR:0.72; 95% died (HR: 0.96; 95% CI: 0.60
Size: 259 patients amiodarone <300 CI:0.530.98; p=0.04) 1.53; p=0.86).
mg/d,
Or addition of This difference was
mexiletine 200 mg TID driven by trends
to amiodarone 300 toward reductions in
mg/d if on rates of appropriate
amiodarone 300 mg/d shocks and episodes
(N=127) of VT storm
VTACH Trial To determine Inclusion Criteria: Patients Study Intervention After 24 mo, 47% of Patients with LVEF >0.30 had
Kuck KH, et whether catheter age 18-80 y with prior MI, ICD plus catheter patients in the greater reduction of VT with
al. 2010 (168) ablation reduces CAD, clinically ablation of all ablation group and catheter ablation than did
20109864 the risk of VT hemodynamically stable VT, inducible VTs or 29% of controls were patients with more severe LV
recurrence in reduced LVEF <0.50, ICD elimination of free of recurrent VT dysfunction (freedom from VT
patients with indication substrate for non- (HR: 0.61;95% CI in 48% with ablation vs 27% of
Ischemic inducible VT 0.370.99, p=0.044). controls, (HR:0.47; 95% CI
Cardiomyopathy, Exclusion Criteria (N=52) 0.240.88, p=0.016).
stable VT, and an MI or Cardiac Surgery within
ICD compared with 1 mo, LV thrombus, artificial Comparator No difference in VT storm,
ICD and continued heart valve, incessant VT, ICD and continued syncope, or death between
medical Rx alone impaired renal function, life medical therapy ablation and controls.
Study Type expectancy <1 y. (N=55)
RCT

89

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study Size
107 patients

CALYPSO Aim Inclusion Criteria Intervention 1 Endpoint Of 243 screened patients, 27


Al-Khatib S. Pilot study to Patients with CAD, ICDs, who Catheter ablation of Mean time to were enrolled.
et al. 2015 determine had received >1 ICD shock or VT (N=13) recurrent VT was 75 d Presently on AAD (88, 41%),
(169) feasibility of RCT of >3 ATP therapies for VT in ablation arm and VT due to reversible cause (23,
25332150 catheter ablation of Comparator 57 d in AAD arm. 11%), and incessant VT (20,
VT vs. AAD when Exclusion Criteria AAD(N=14) 9%).
used early in the Present AAD, Incessant VT, There were 2 deaths
course of patients VT due to reversible cause in both arms of the
with CAD who study
experience ICD
therapies.
Study Type
Pilot RCT
Study size
27 patients

Data Supplement 23. Nonrandomized Trials, Observational Studies, and/or Registries of Treatment of Recurrent Arrhythmias in IHD
(Section 7.1.3)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Conclusions
Author; Study Size (P values; OR or RR;
Year Published & 95% CI)
Blanck et al. Study type: Inclusion criteria: Results: BBRVT typically occurs in patients
1993 (170) Single Center Review All patients at single center 45 of 48 patients had SHD with SHD from a variety of causes in
8269297 with BBRVT diagnosed at EPS SHD was NICM in 16 patients, patients with prolonged HV
Size: 48 patients between 1980-1992 ischemic cardiomyopathyin 23 conduction intervals.
Criteria: patients, V HD in 2 patients
1) Typical RBBB or LBBB BBRVT is associated with aborted
QRS morphology Mean LVEF 23.2% SCD, Syncope, and Palpitations
during VT

90

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
2) QRS preceded by His Clinical Presentation BBRVT is most commonly
and appropriate Aborted SCD in 26% associated with a LBBB QRS
bundle branch Syncope in 51% morphology, and less commonly
potential Sustained palpitations in 10% with RBBB or Interfascicular QRS
3) Stable HV, RB-V, or morphologies
LB-V interval Mean HV interval in sinus 80.4
4) Induction dependent msec Catheter ablation targeting the
on HV delay RBB or LBB is highly effective and
5) Termination by block QRS morphology in VT associated with a low risk of serious
in HPS LBBB in 46 patients complications.
6) Noninducibility after RBBB in 5 patients
RBB ablation Interfascicular reentry in 2 patients

Catheter Ablation
Performed in 28 patients targeting
the RBB in 26 patients and LBB in 2
patients
Successful ablation of VT in 100%
No Complications observed.
Brugada J et al. Study type: Inclusion: prior MI, Results: 61 patients were inducible In patients with VA in the chronic
2001 (171) prospective spontaneous VA not related to into sustained VA. phase of MI, probability of
11216974 an acute ischemic event and recurrence is high despite coronary
Size: 61 patients coronary lesions requiring After revascularization, 52 of 59 artery revascularization, but
revascularization patients previously inducible were mortality is low if combined with
still inducible (group A), and 10 appropriate AAD.
Exclusion: n/a patients were noninducible (group
B). Recurrences: lower EF predicted
Protocol: EP performed before higher recurrence rate but not
and after revascularization No differences were found in ischemia before revascularization,
clinical, hemodynamic, therapeutic amiodarone or BB therapy or EP
and electrophysiological study after revascularization. An EF
characteristics between both <30% predicted recurrent
groups. arrhythmic events (p=0.02), but not
the presence of demonstrable
During 32 +/- 26 mo followup, ischemia before revascularization
28/52 patients in group A (54%) (p=0.42), amiodarone (p=0.69) or

91

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
and 4/10 patients in group B (40%) beta-adrenergic blocking agent
had arrhythmic events (p =0.46). therapy (p=0.53).

Total mortality was 10% in both


groups.
Sears et al. 1999 Study type: literature Inclusion: studies assessing Results: 13-38% of recipients Conclusions: Psychosocial
(172) review psychological impact of ICD experiencing diagnosable levels of adjustment risk profiles indicate that
10410293 and shocks anxiety. young ICD recipients and those with
high discharge rates may experience
Specific ICD-related concerns such the most adjustment difficulties
as fear of shock, fear of device
malfunction, fear of death, and fear
of embarrassment have been
identified.

Lopera et al. Study type: Inclusion criteria: Results: BBRVT occurs in patients with
2004 (173) Single Center Review His Bundle, LBB, or RBB HPS VT induced in 20 of 234 both NICM and ischemic
15028072 potential closely associated consecutive patients referred for cardiomyopathy, usually with
Size: 20 patients with QRS with any of VT ablation impaired LVEF.
the following:
1) H-H interval variation NICM: 9 of 81 patients (11%) had BBRVT is most commonly
preceding similar V-V HPS VT associated with a LBBB QRS
interval variation; ICM: 11 of 153 patients (7.1%) had morphology, and less commonly
2) Anterograde HPS VT with RBBB or Interfascicular QRS
activation of the Mean LVEF 29+17% morphologies
bundle branches 2 of 20 patients had normal LVEF
during tachycardia; Catheter ablation targeting the
or, Clinical Presentation RBB or LBB is highly effective and
3) Abolition of VT by ICD Shocks in 10 patients associated with a low risk of serious
bundle branch Syncope in 3 patients complications if only one BB is
ablation. Other symptoms in 7 patients targeted and a higher risk of AV
block if both BBs are targeted for
Exclusion criteria: None Typical BBRVT in 16 of 20 patients ablation.
(all had LBBB QRS morphology)
13 of 16 patients BBRVT
successfully ablated by RBB

92

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
ablation and 3 of 16 by LBB
ablation.
HV interval prolonged from 70+5.9
msec to 83+17 msec after ablation.

Typical BBRVT and Interfascicular


VT in 2 of 20 patients. Ablation of
both the RBB and portion of LBB
eliminated VT in both patients,
complicated by AV block in 1 pt.

Focal Mechanism from BBs in 2


patients, one in RBB, one in LBB.
Ablation eliminated focal VT in
both patients, complicated by AV
block in 1 pt.
Mehdirad et Study type: Inclusion criteria: Results: Catheter ablation of the RBB is
al.1995 (174) Single Center Review All patients undergoing RF HV interval 68+8 msec at baseline effective for the treatment of BBRVT
8771124 catheter ablation of the RBB LVEF mean 31+15%
Size: 16 patients for BBRVT BBRVT is associated with
RBBB developed in 15/16 patients prolonged HV conduction intervals.
after RBB ablation
AV block occurred in 1 pt The medium term followup after
After mean of 1910 mo, one catheter ablation of the RBB is
patient died suddenly, 2 received overall quite good.
cardiac transplantation, and 1 died
of CHF.
HELP-VT Aim: Inclusion criteria: 1 endpoint: At 1 y follow-up, VT Complications
Dinov B, et al. To determine the Patients with SHD referred for free survival was 57% for ischemic Complications occurred in 11.1% of
2014 (175) outcome of VT catheter ablation of VT with cardiomyopathyand 40.5% for NICM and 11.1% of ischemic
24211823 catheter ablation in either NICM (N=63) or NICM patients (HR: 1.62; 95% CI cardiomyopathypatients, including
patients with NICM to ischemic 1.122.34, p=0.01). ischemic death in 4.8% of NICM and 3.7% of
those with ICM cardiomyopathy(N=164) cardiomyopathyrequired epicardial ischemic cardiomyopathy
Study type: ablation in only 2 of 164 (1.2%)
Prospective, non- Exclusion criteria: whereas NICM required epicardial
randomized Failure of informed consent ablation in 30.8% (p=0.0001).
Size: 227 patients

93

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Intervention:
Catheter ablation for patients
with NICM

Comparator:
Catheter ablation in patients
with ICM
Euro-VT Study Aim Inclusion Criteria 1 endpoint: Complications
Tanner H 2010 To determine the Drug and device refractory, Acute success with ablation was Major complications occurred in
(176) safety and efficacy of recurrent sustained VT after achieved in 83% of mappable VTs 1.5% and minor complications in 5%
19656251 electroanatomic MI. and 40% of non-mappable VTs of patients, particularly groin
mapping and irrigated >4 episodes of sustained VT in (p<0.0001). hematomas, with no procedural
RF catheter ablation prior 6 mo. deaths.
for VT after MI During 12mo follow-up, VT
Exclusion Criteria recurred in 49% of patients.
Study Type: Age <18 y
Multicenter, non- MI within 2 mo The mean number of therapies
randomized LV Thrombus dropped from 6070 prior to
Unstable Angina ablation to 1415 in the same
Study Size Severe AS or MR period of time (6 mo) after ablation
63 patients Unwillingness to participate (p= 0.02).

Intervention
Electroanatomic mapping and
ablation with open-tip
irrigated catheter.
Post-approval Aim Inclusion Criteria 1 endpoint: Comments
Thermocool Trial To evaluate long-term Patient with coronary disease, At 6 mo: 62% without VT Reduction in amiodarone usage
Marchlinski F safety and age 18 y and LVEF 10% with recurrence, proportion of patients and hospitalization
2016 (177) effectiveness of RF recurrent VT (either 4 with ICD shock reduced from 81.2 Improvement in QoL
26868693 catheter ablation for episode documented by ICD, (pre) to 26.8% and 50% reduction
VT in patients with 2 episode documented by in VT episodes in 63.8% of patients.
CAD ECG in patients without ICD,
incessant VT or symptomatic Safety Endpoint
Study Type: VT despite AAD treatment CV specific AE in 3.9% with no
Multicenter, non- stroke
randomized Exclusion Criteria

94

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Mobile LV thrombus, MI
Study Size within 3 mo, idiopathic VT,
249 patients class IV HF, creatinine 2.5,
recent cardiac surgery,
unstable angina, severe AS or
MR.

Intervention
Electroanatomic mapping and
ablation with open-tip
irrigated catheter.
International Aim: to determine Inclusion criteria: SHD with 1 endpoint: Procedural complications occurred
VT Collaborative the association of VT ischemic and non-ischemic Freedom from VT recurrence, Heart in 6%, including 2 deaths (0.1%),
Group Study recurrence after cardiomyopathies with Transplant, or death was 70% at 1 y hemopericardium in 1.7%, and
Tung R 2015 ablation and survival monomorphic VT and follow-up. vascular access complications in
(178) in scar related VT myocardial scar by VT recurred in 55% of patients who 1.6%
26031376 electroanatomic mapping died vs. 22% of patients who
Study type: survived.
Multicenter Exclusion criteria: absence of Transplant free survival was 90%
observational scar on electroanatomical for patients without VT recurrence
mapping and 71% for those with VT
Size: 2061 recurrence (HR: 6.9; 95% CI: 5.3
Intervention: Catheter 9.0, p<0.001).
ablation, either endocardial or
epicardial, guided by EAM.
End point of ablation with
elimination of all induced VTs
Meta-Analysis Aim: To determine PubMed, Embase, Cochrane 1 endpoint: Electrical Storm occurred in 17 of
of Randomized the relative risk of VT searches of both randomized VT recurred in 93 of 266 patients 116 (15%) after catheter ablation
and Non- recurrence in patients and nonrandomized clinical (35%) after Catheter Ablation and 29 of 119 (25%) on AAD therapy
Randomized undergoing catheter trials of catheter ablation of VT compared with 105 of 191 (55%) on (HR: 0.61; 95% CI: 0.361.03,
Trials of Catheter ablation compared compared with a control group AAD (HR: 0.62; 95% CI: 0.510.76, p<0.066).
Ablation for VT with medical therapy receiving AAD treatment alone p<0.001)
Mallidi J 2011 Mortality occurred in 12% of
(179) Study type: Intervention: Safety endpoint: patients treated with ablation and
21147263 Meta-Analysis of 5 Catheter ablation with or Complications occurred in 6.3% 14% on AAD.
Trials of VT Ablation without AAD after ablation, including death

95

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
(1%), tamponade (1%) and AV block
Size: Comparator: (1.6%)
457 patients AAD alone.

Cooled Tip Aim: Inclusion criteria: 1 endpoint: Complications


Ablation of VT To determine the >2 episodes of Acute success with elimination of Complications occurred in 8%
Calkins 2000 safety and efficacy of hemodynamically stable VT in all mappable VTs in 75%, including death in 2.7%
(180) an internally cooled previous 2 mo, CAD, ICD
10841242 RF ablation catheter implantation, failure of >2 At a mean of 243+153 d of follow-
used for VT in SHD in AAD. up, VT recurred in 46% of patients
patients with >2
episodes of VT in the Exclusion criteria: Acute success defined by
prior 2 mo despite >2 Failure to give informed noninduciblity of VT after ablation
AAD consent did not predict VT recurrence

Study type: Intervention:


Non-Randomized trial Catheter ablation using the
of Cooled Tip ablation Cooled RF catheter system
catheter for VT Comparator:
VT recurrence Hx prior to
Size: ablation
147 patients
Multicenter Aim: Inclusion criteria: 1 endpoint: 1 y mortality was 18%
ThermoCool To determine the >4 episodes of sustained VT Freedom from recurrent VT at 6 mo
Ventricular outcome after requiring cardioversion or AAD follow-up in 123/231 patients
Tachycardia catheter ablation of for termination in past 6 mo (53%).
Ablation Trial VT despite ICD or AAD THERAPY,
Stevenson WG, age >18 y. VT ablation reduced the median
et al. 2008 (181) Study type: number of VT episodes in 6 mo
19064682 Non-randomized Exclusion criteria: before ablation from 11.5 to 0 after
LVEF <0.10, LV thrombus, ablation (p<0.0001)
Size: Creatinine >2.5, NYHA Class IV
231 patients CHF, severe AS, unstable Safety endpoint:
angina, pregnancy. Complications occurred in 7%,
including 7 patients (3%) who died
Intervention: within 3 d of ablation, and groin
complications in 4.7%.
96

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Catheter ablation with the
BioSense ThermoCool ablation
catheter
Comparator:
Prior Hx of VT recurrences
Steinberg et al. Study type: cohort Patient with sustained post- 1 endpoint: 12 patients (3.1%) Results (cont.): Patients with VT
1999 (150) study operational VT 24 h but <30 d experienced 1 episode of were more likely to have prior MI
10027813 after CABG among consecutive sustained VT 4.14.8 d after CABG (92% vs. 50%, p<0.01), severe CHF
Size: 12 patients patients 382 patients (56% vs. 21%, p<0.01), and LVEF
undergoing CABG at a single In 11 /12 patients, no <0.40 (70% vs. 29%, p<0.01).
institution postoperative complication By multivariate analysis, the
explained the VT. 1 patient had a number of bypass grafts across a
Variables associated with the perioperative MI. noncollateralized occluded vessel to
occurrence of VT was an infarct zone was the only
performed The in-hospital mortality rate was independent factor predicting VT.
25%. Among the 9 survivors, 5 had Conclusions: (1) Patients who
EPS with all inducible sustained developed VT had a high in-hospital
monomorphic VT (matching clinical mortality rate of 25% (2) However,
VT). 3/9 patients received an ICD long-term outcome was good
before hospital discharge. Other (possibly related to antiarrhythmic
6/9 patients received chronic or ICD). (3) predictors are MMVT
therapy with AAD (primarily previous MI scar and associated
amiodarone). severe LV dysfunction. (4)
Relationship was found between the
All 9 patients are alive, with a mean development of VT and the
follow-up of 2.5 y. placement of a bypass graft across a
noncollateralized occluded coronary
2 patients (1 with an ICD and 1 on vessel to a chronic infarct zone. (5)
amiodarone) had recurrent VT The development of MMVT was
during followup. typically not due to a detectable
postoperative complication or
ischemia.

97

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 24. Nonrandomized Trials, Observational Studies, and/or Registries of NICM (Section 7.2)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Ackerman MJ 2011 Study type: HRS/EHRA Expert consensus General: Class I: 1) sound clinical LQTS: Note difference between
(182) consensus statement. statement on the state of suspicion when positive Class I if QTc >480 or 500 ms, and
21810866 genetic testing for the predictive value > 40%, Class IIb if QTc >460/480 ms
channelopathies and signal/noise ratio >10; 2) AND/OR
cardiomyopathies genetic test result provides either
diagnostic or prognostic info, or
Panel: geneticists, influences therapeutic choices.
arrhythmia specialists Screening of family members:
Agreement 84% when genetic testing leads to the
adoption of therapy/protective
measures/ lifestyle adaptations.

LQTS: Class I: 1) any pt with


strong clinical index of suspicion
for LQTS; 2) any asymptomatic pt
with QT prolongation on serial
ECGs: QTc >480 ms prepuberty;
>500 ms, adult; 3) Mutation
specific genetic testing for family
members and other appropriate
relatives
Class IIb: any asymptomatic pt
with otherwise idiopathic QTc
values >460 ms (puberty) or 480
ms (183) on serial ECGs

CPVT: Class I: 1) any pt w strong


clinical index of suspicion of
CPVT;

98

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
2) Mutation specific genetic
testing is recommended for
family members and appropriate
relatives

Brugada: Class I: Mutation


specific genetic testing is
recommended for family
members and appropriate
relatives
Class IIa: any pt w strong clinical
index of suspicion of BrS,
including with procainamide
challenge
Class III: not indicated in the
setting of an isolated type 2 or 3
Brugada ECG pattern

Short QTS: Class I: Mutation


specific genetic testing is
recommended for family
members and appropriate
relatives
Class IIb: any pt with strong
clinical index of suspicion

ARVC: Class I: Mutation specific


genetic testing is recommended
for family members and
appropriate relatives
Class IIa: can be useful for
patients satisfying task force
diagnostic criteria
Class IIb: may be considered for
patients with possible ACM/ARVC
Class III: not recommended for
patients with only a single minor

99

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
criterion according to the 2010
task force criteria

SCD/SIDS: Class I: 1) Collection of


tissue sample recommended
(blood or heart/liver/spleen
tissue); 2) Mutation specific
genetic testing is recommended
for family members and
appropriate relatives
Class IIb: testing may be
considered if circumstantial
evidence suggests LQTS or CPVT
specifically

ACA/resuscitated: Class I:
Genetic testing should be guided
by the results of medical
evaluation and is used for the 1
purpose of screening at-risk
family members for sub-clinical
disease
Class III: Routine genetic testing,
in the absence of a clinical index
of suspicion for a specific
cardiomyopathy or
channelopathy, is not indicated
for the survivor of unexplained
OHCA

HCM: Class I: 1) any pt in whom


the clinical dx of HCM is
established. 2) Mutation specific
genetic testing is recommended
for family members and
appropriate relatives

100

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
DCM: Class I: 1) DCM and
significant cardiac conduction
disease and/or family Hx of
premature unexpected sudden
death. 2) Mutation specific
genetic testing is recommended
for family members and
appropriate relatives

LVNC: Class I: Mutation specific


genetic testing is recommended
for family members and
appropriate relatives
Class IIa: can be useful if clinical
dx of LVNC is established

PCCD: Class I: Mutation specific


genetic testing is recommended
for family members and
appropriate relatives
Class IIb: may be considered as
part of diagnostic evaluation for
patients with either isolated CCD
or CCD with concomitant
congenital heart disease,
especially w post family Hx of
CCD.

Hershberger RE et al. Study type: This is a N/A N/A Idiopathic DCM, has been shown to
2010 (184) review on clinical and have a familial basis in 20-35% of
20864896 genetic issues in DCM cases. Genetic studies in familial
dilated cardiomyopathy have shown
dramatic locus heterogeneity with
mutations identified in >30 mostly
autosomal genes showing primarily
dominant transmission.

101

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Piers et al 2013 (185) Study type: single center, Inclusion criteria: 1 endpoint: VT recurrence over VT recurrence is high in NICM
24036134 observational Patients with NICM and mean follow up of 2515 mo patients, but significant reduction in
VT treated with catheter the frequency of VT episodes is
Size: 45 ablation Results: VT occurred in 24 observed in the majority of patients
patients (53%), but the 6 mo VT following ablation.
Exclusion criteria: N/A burden was reduced by 75% in
79%. There was a suggestion that
Recurrence rates were low after patients treated with ablation early
complete procedural success (first VT or VT ICD therapy) had better
(18%), but high after both partial outcome than those treated late.
success (77%) and failure (73%).
Greulich et al. 2013 Aim: study aimed to Inclusion criteria: 155 1 endpoint: 1 endpoints were Could not tell on additional LGE
(186) demonstrate that the consecutive patients with death, aborted SCD, and parameters due to low numbers.
23498675 presence of late systemic sarcoidosis who appropriate ICD discharge.
gadolinium enhancement underwent CMR for
is a predictor of death workup of suspected Results: LGE was present in 39
and other adverse events cardiac sarcoid patients (25.5%). The presence of
in patients with involvement. The median LGE yields a HR of 31.6 for death,
suspected CS follow-up time was 2.6 y. aborted SCD, or appropriate ICD
discharge, and of 33.9 for any
Study type: Multicenter Exclusion criteria: N/A event. This is superior to
prospective functional or clinical parameters
such as left LVEF, LV end-diastolic
Size: 155 patients volume, or presentation as HF,
yielding HRs between 0.99 (per %
increase LVEF) and 1.004
(presentation as HF), and
between 0.94 and 1.2 for
potentially lethal or other
adverse events, respectively.
Kuruvilla et al. 2014 Aim: To assess the Inclusion criteria: NICM 1 endpoint: Patients with LGE Patients with LGE had increased
(187) relation between CMR had an increased risk of SCA overall mortality (OR: 3.27;
24363358 LGE and cardiovascular Exclusion criteria: events (OR: 5.32; p<0.00001) p<0.00001) and increased HF
outcomes in NICM Ischemic cardiomyopathy, compared with those without hospitalization (OR: 2.91; p=0.02),
patients HCM LGE. The annualized event rates for SCA
Intervention: CMR-LGE was 6.0% in LGE detected patients vs.
findings and subsequent 1.2% for those without LGE (p<0.001).

102

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: Meta- clinical outcomes in
Analysis patients with NICM

Size: 9 studies and 1,488 Comparator: N/A


patients
HELP-VT Study type: single center, Inclusion criteria: 1 endpoint: VT free survival at 1 VT free survival worse in NICM
Dinov B et al. 2014 observational Patients with SHD y compared to ICM.
(175) referred for catheter
24211823 Size: 227 (63 NICM) ablation of VT with either Results: VT free survival 40.5% in Complete noninducibility after
NICM (N=63) or ischemic NICM vs. 57% in ICM index procedure predicted better
cardiomyopathy (N=164) outcome
HR for VT recurrence for NICM
Exclusion criteria: 1.62 (p=0.01)
Failure of informed
consent
Tokuda et al 2012 Study type: single Inclusion criteria: 1 endpoint: All cause death or Outcomes of ablation differ in
(188) center, observational Patients with NICM and heart transplantation following individual etiologies of NICM. ARVC
22942218 sustained monomorphic ablation; 2 endpoint: composite had better outcomes than DCM for 1
Size: 226 VT referred for catheter of death, heart transplantation (p=0.002) and 2 end points (p=0.004).
ablation and admission for VT recurrence Sarcoidosis had worse outcome than
DCM for 2 end point (p=0.002).
Exclusion criteria: N/A Results: After a mean of 1.4
ablation procedures
1 endpoint (4.43.3 y follow-up)
reached in 66 (29%) patients
reached the 1 end point: death
in 50 (21%) and transplant in 16
(7%)

2 endpoint (12 mo): death 10%,


transplant 3%, VT admission 18%

Cantero-Prez EM, et Aim: To evaluate the Inclusion criteria: Results: Appropriate ICD therapies were
al. 2013 (155) effectiveness of ICDs for Records from patients Median follow-up of 77 d recorded in 42.9% (12/28) in this
24314988 primary prevention in accepted for heart overall mortality in the ICD group population.
patients with LVEF 30% transplantation from was 7.1% (2/28) and in the non-
January 1, 2006, to July

103

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
included on the heart 30, 2012, and whose LVEF ICD group was 17.6% (9/51;
transplantation list was <31% were reviewed p=0.062).
Cause of death in patients
Size: Patients who without ICDs:
received ICDs for primary Sudden death (5/9, 55.6%),
prevention (N=28) were HF (4/9, 44.4%).
compared with patients Cause of death in patients with
without ICDs (N=51) ICDs: HFheart
Frhlich GM, et al. Aim: To delineate the Inclusion criteria: Results: ICDs appear to be associated with a
2013 (156) role of ICD therapy for Patients listed for heart Median time on the waiting list = reduction in all-cause mortality in
23813845 the primary and transplantation in 2 8 mo (estimated 1-year: 883% patients implanted with the device for
secondary prevention of tertiary heart transplant vs. 773% vs. 673%; p=0.0001). primary and secondary prevention
SCD in patients listed for centres were enrolled. Of An independent beneficial effect compared to those without an ICD.
heart transplantation 550 patients (51%) on the of ICDs that was most
transplant list with an pronounced in patients who had
Size: N=1089 ICD: received an ICD for primary
primary prevention ICD: prevention (HR: 0.4, 95% CI:
N=216 0.190.85; p=0.016).
secondary prevention
ICD: N=334
Gandjbakhch E, et al. Aim: To evaluate the ICD Inclusion criteria: Results: Need for mechanical circulatory
2016 (157) benefit on mortality in Patients with end-stage 15.6% of patients died while support (p<0.001), low EF (p=0.001)
27344378 patients with end-stage HF receiving an ICD awaiting heart transplantation. and registration on the regular list
HF listed for heart before or within 3 mo Non-ICD patients presented more (p=0.008) were the only independent
transplantation after being listed for often haemodynamic predictors of death.
heart transplantation compromise. ICD-related complications occurred
Size: N=380 consecutive ICD did not remain an in 21.4% of patients, mainly as a result
patients listed for heart independent predictor of death. of postoperative worsening of HF
transplantation between Death by haemodynamic (11.9%).
2005 and 2009 in A compromise (76.3% of deaths),
tertiary heart transplant which occurred more frequently
centre in the non-ICD group (14.7% vs.
5.8%; log-rank p=0.002).

Unknown/arrhythmic deaths did


not differ significantly between

104

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
the two groups (3.9% vs. 1.7%;
log-rank p=0.21).
Vakil K, et al. 2016 Aim: To assess the Inclusion criteria: Results: In the subgroup of patients with
(158) impact of ICD on waitlist Adults (age 18 y) listed Median follow-up of 154 days, LVAD (N= 9,478), having an ICD was
mortality in patients for first-time heart 3,638 patients (11%) died on the associated with an adjusted 19%
listed for heart transplantation in the US waitlist (9% in ICD group vs. 15% relative reduction in mortality (HR:
transplantation between January 1, 1999, in no-ICD group; p<0.0001), 0.81; 95% CI: 0.700.94).
and September 30, 2014, whereas 63% underwent heart
Size: N=32,599 were retrospectively transplantation.
identified from the An ICD at listing was associated
United Network for Organ with an adjusted 13% relative
Sharing registry. reduction in mortality (HR: 0.87;
95% CI: 0.800.94).
Oloriz et al 2014 Study type: single Inclusion criteria: 1 endpoint: VT recurrence, Multivariate predictors of VT
(189) center, observational Patients with NICM and stratified to scar location recurrence included electrical storm
24785410 drug refractory VT (anteroseptal vs. basal lateral) (HR: 3.211; p=0.001) and NHYA class
Size: 87 treated with ablation determined by unipolar voltage (HR: 1.608; p=0.018), anteroseptal
mapping scar pattern (HR: 5.547; p<0.001)
Exclusion criteria: N/A
Results: Over a mean 1.5 y
follow up, VT recurred in 44
patients (51%) during a median
follow-up of 1.5 y. Anteroseptal
scar was associated with higher
VT recurrence (74% vs. 25%; log-
rank p<0.001)

Death occurred in 15%

Proietti et al 2015 Study type: single center, Inclusion criteria: 1 endpoint: VT recurrence, Results of substrate guided ablation
(190) observational Patients with ischemic determined by ICD interrogations less favorable in NICM than ischemic
25488957 cardiomyopathyand over 641301 d. cardiomyopathy patients
Size: 142 (55 NICM) NICM referred for
catheter ablation for VT Results: Recurrent VT occurred
more frequently in the NICM
Exclusion criteria: N/A group 51% than in the ischemic

105

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
cardiomyopathy group 26%
(p=0.03)

Acute results (defined by


response to PES) correlated with
likelihood of recurrence: for the
NICM group, recurrence was
observed in 7, 75 and 100% of
successful, partially successful
and failed ablations
Haqqani et al 2011 Study type: single center, Inclusion criteria: 1 endpoint: VT recurrence over Isolated septal substrate in NICM
(191) observational Patients with NICM and mean followup of 2028 mo portended a poor outcome, both in
21392586 VT treated with catheter terms of VT recurrence and transplant
Size: 31 ablation who had isolated Results: Following a mean of 1.6 free survival in followup
intra-septal scar (11.65% ablation procedures, VT
of total) recurrence was observed in 32%;
death and heart transplant
Exclusion criteria: N/A occurred in 26% and 16%
respectively
Kuhne et al 2010 Study type: single center, Inclusion criteria: 1 endpoint: VT recurrence over
(192) observational Patients with NICM and mean followup of 1813 mo
20384656 VT treated with catheter
Size: 35 ablation Results: Recurrence was
observed in 57%. In patients who
Exclusion criteria: N/A had isolated late potentials
(targeted for ablation), freedom
from VT and major arrhythmia
related adverse events was
improved compared to those
without identified isolated late
potentials
Cano et al 2009 (193) Study type: single center, Inclusion criteria: 1 endpoint: VT recurrence over The VT substrate in NICM is often
19695457 observational Patients with NICM and mean follow up of 187 mo more prominent on the epicardial
VT suspected to be following endocardial and than the endocardial surface.
Size: 22 epicardial in origin (Prior epicardial ablation Epicardial ablation may improve
failed endocardial outcome in selected patients with VT
in the setting of NICM.

106

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
ablation or ECG Results: Freedom from VT
characteristics during VT) recurrence was observed in 15 of
21 patients in whom any ablation
Exclusion criteria: N/A was performed, and 14 of 18 with
epicardial ablation
Delacretaz et al 2000 Study type: single center, Inclusion criteria: 1 endpoint: VT recurrence over Recurrent monomorphic VT in NICM
(194) observational Patients with NICM and mean followup of 1512 mo can be focal or reentrant; reentrant
10695454 VT treated with catheter causes can be scar related or 2 to
Size: 26 ablation Results: VT recurrence was bundle branch reentry.
observed in 23%, but differed
Exclusion criteria: N/A depending on VT mechanism: 40,
0 and 14% in scar related VT,
focal VT and bundle branch
reentry, respectively.

Data Supplement 25. RCTs Secondary Prevention SCD in NICM (Section 7.2.1)
Study Acronym; Aim of Study; Patient Population Endpoint Results Relevant 2 Endpoint (if
Author; Study Type; (Absolute Event Rates, any);
Year Published Study Size (N) P values; OR or RR; & Study Limitations;
95% CI) Adverse Events
AVID Aim: To examine Inclusion criteria: patients who were 1 endpoint: Survival Study terminated
The AVID the effect on overall resuscitated from near-fatal VF; sustained early after 1016 of 1200
Investigators survival of initial VT with syncope; or sustained VT with an Results: Overall survival was greater patients enrolled
1997 (131) therapy with an ICD LVEF 0.40 and symptoms suggesting with the ICD, with unadjusted 81% of patients had
9411221 as compared with severe hemodynamic compromise. estimates of 89.3 percent, as CAD
amiodarone or compared with 82.3% in the AAD
sotalol in patients Exclusion criteria: arrhythmia was judged group at 1 y, 81.6% vs 74.7% at 2 y,
resuscitated from VF to have a transient or correctable cause, and 75.4% vs 64.1% at 3 y (p<0.02).
or symptomatic, excessively high risk (life expectancy <l y, The corresponding reductions in
sustained VT with class IV CHF, awaiting a heart transplant, or mortality (with 95% confidence
hemodynamic requiring a balloon pump, other mechanical limits) with the ICD were 3920%,
compromise. means, or inotropic drug administration for 2721%, and 3121%.
hemodynamic support)
Study type: RCT or excessively low risk (event occurring
within 5 d of cardiac surgery or angioplasty,
107

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 1016 patients or occurring in-hospital within 5 d after MI),
had a previous ICD implant (or attempted
implant), chronic serious bacterial infection,
or were unable to give verbal assent due to
neurologic impairment. Contraindications
to amiodarone.

Intervention: Therapy with ICD

Comparator: AAD amiodarone or sotalol,


but only 2.6% received sotalol, most
received amiodarone
CIDS Aim: To compare Inclusion criteria: in the absence of either 1 endpoint: Death from any cause. 82% had ischemic
Conolly et al. the efficacy of the recent AMI or electrolyte imbalance, they Results: A nonsignificant reduction etiology
2000 (132) ICD and amiodarone manifested any of the following: (1) in the risk of death was observed
10725290 for the prevention documented VF; (2) OHCA requiring with the ICD, from 10.2%/y to
of death in patients defibrillation or cardioversion; (3) 8.3%/y (RRR: 19.7; 95% CI: -7.7%
with previous documented, sustained VT causing syncope; 40%; p=0.142). A nonsignificant
sustained (4) other documented, sustained VT at a reduction in the risk of arrhythmic
ventricular rate 150 beats/min, causing presyncope or death was observed, from 4.5%/y to
arrhythmia angina in a patient with a LVEF 35%; or (5) 3.0%/y (RRR :32.8%; 95% CI: -7.2%
unmonitored syncope with subsequent 57.8%; p=0.094).
Study type: RCT documentation of either spontaneous VT
10 s or sustained (30 s) monomorphic VT
Size: 659 patients induced by programmed ventricular
stimulation.

Exclusion criteria: (1) ICD or amiodarone


not considered appropriate, (2) excessive
perioperative risk for ICD implantation; (3)
previous amiodarone therapy for 6 wk; (4)
nonarrhythmic medical condition making 1
y survival unlikely, and (5) LQTS.

Intervention: ICD

Comparator: Amiodarone

108

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
CASH Aim: to study the Inclusion criteria: patients resuscitated 1 endpoint: The 1 end point was In ICD patients, the
Kuck et al. 2000 impact on overall from CA 2 to documented sustained VA all-cause mortality. percent reductions in
(133) survival of initial Results: Over a mean follow-up of all-cause mortality were
10942742 therapy with an ICD Exclusion criteria: If CA occurred within 72 5734 mo, the death rates were 41.9%, 39.3%, 28.4%,
as compared with h of an AMI, cardiac surgery, electrolyte 36.4% (95% CI: 26.9%46.6%) in the 27.7%, 22.8%, 11.4%,
that with 3 AAD. abnormalities, or proarrhythmic drug ICD and 44.4% (95% CI: 37.2% 9.1%, 10.6%, and 24.7%
effect. 51.8%) in the at 1 y to 9 of follow-up.
Study type: RCT amiodarone/metoprolol arm. CAD was etiology in
Intervention: ICD therapy Overall survival was higher, though 73%
Size: 288 patients not significantly, in patients assigned A much larger
Comparator: amiodarone, metoprolol, or to ICD than in those assigned to drug reduction of 61%, for
propafenone. Assignment to propafenone therapy (HR: 0.766, 97.5% CI:1.112, SCD was observed
was in March 1992, after an interim analysis p=0.081).
showed a 61% higher all-cause mortality
rate than in 61 ICD patients during a
followup of 11.3 mo.
Desai et al. Aim: To determine Inclusion criteria: prospective RCT of ICD 1 endpoint: Two of the 3 2 Analysis of all 7 trials
2004 (195) whether ICD therapy or combined CRT defibrillator vs medical prevention trials presented (1 and 2 prevention)
15598919 reduces all-cause therapy enrolling at least some individuals subgroup estimates for ICD efficacy combined demonstrated
mortality in patients with NICM and reporting all-cause mortality in NICM. Pooled analysis of these 2 a statistically significant
with NICM. as an outcome. prevention trials (N=256 patients 31% overall reduction in
with NICM) indicated an equivalent mortality with ICD
Study type: meta- Intervention: ICD to 1 y prevention but nonsignificant therapy (RR: 0.69; 95%
analysis of RCT mortality reduction with ICD therapy CI: 0.560.86; p=0.002).
Comparator: Medical therapy. (RR: 0.69; 95% CI: 0.391.24;
Size: 8 randomized p=0.22).
trials enrolling a
total of 2146
patients with NICM
were included.
MAVERIC Aim: to test the Inclusion criteria: survivors of sustained 1 endpoint: Survival and 61% of patients had
Lau et al. 2004 possibility of VT, VF or sudden cardiac death in the arrhythmia recurrence prior MI
(135) prospectively absence of an AMI in the last 48h. EPS has a minimal
15172648 identifying patients Results: Of the 108 EP arm patients, impact on the diagnosis
who would benefit Exclusion criteria: life expectancy of <6 mo 31 (29%) received an ICD, 46 (43%) of patients presented
most ICD by EPS in from a non-arrhythmic cause or child- received AAD only (mainly with VT, VF or SCD.
bearing age amiodarone or sotalol) and 18 (17%)

109

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
the context of 2 Intervention: EP-guided interventions received coronary revascularization The trial does not
prevention. (AAD, coronary revascularization, and ICD) but no ICD. No significant support a role for EP
(106 patients assigned to this arm) differences in survival or arrhythmia testing in risk
Study type: RCT recurrence existed between the 2 stratification.
Comparator: therapy with amiodarone (108 treatment arms after 6 y. However,
Size: 214 patients patients assigned to this arm) ICD recipients had a lower mortality
than non-ICD recipients, regardless
of allocated treatment (HR:0.54,
p=0.0391).
Claro et al. Aim: To evaluate Inclusion criteria: Randomised and quasi- 1 endpoint: SCD and overall For 2 prevention, the
2015 (136) the effectiveness of randomised trials assessing the efficacy of mortality quality of the evidence
26646017 amiodarone for 1 amiodarone vs. placebo, no intervention, or was very low, so the
or 2 prevention of other antiarrhythmics in adults, either for Results: For 2 prevention, authors concluded that
SCD compared with 1 prevention or 2 prevention of SCD. amiodarone compared to placebo or there was uncertainty
placebo or no no intervention (2 studies, 440 on the findings.
intervention or any Exclusion criteria: NA participants) appeared to increase Amiodarone was
other Intervention: Amiodarone the risk of SCD (RR: 4.32; 95% CI: associated with an
antiarrhythmic. 0.8721.49) and all-cause mortality increase in pulmonary
Comparator: placebo, no intervention, or (RR:3.05;95% CI 1.337.01). and thyroid adverse
Study type: meta- other antiarrhythmics However, the quality of the events.
analyses using a evidence was very low. Compared to
random-effects other antiarrhythmics (4 studies,
model 839 participants) amiodarone
appeared to increase the risk of SCD
Size: 24 studies (RR:1.40; 95% CI: 0.563.52; very
(9,997 participants) low quality of evidence), but there
was no effect in all-cause mortality
(RR: 1.03; 95% CI: 0.751.42; low
quality evidence).
OPTIC Study Aim: To determine Inclusion criteria: Patients were eligible if 1 endpoint: ICD shock for any Amiodarone plus BB is
Connolly et al. whether they had received an ICD within 21 d for reason. effective for preventing
2006 (159) amiodarone plus BB inducible or spontaneously occurring VT or these shocks and is
16403928 or sotalol are better VF. Results: Shocks occurred in 41 more effective than
than BB alone for patients (38.5%) assigned to BB sotalol but has an
prevention of ICD Exclusion criteria: Patients were excluded if alone, 26 (24.3%) assigned to increased risk of drug-
shocks. they had LQTS, corrected QT interval of sotalol, and 12 (10.3%) assigned to related adverse effects
more than 450 millisec, were receiving a amiodarone plus BB. A reduction in

110

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: class I or class III antiarrhythmic agent, had the risk of shock was observed with Adverse pulmonary
multicenter RCT received amiodarone or sotalol for more use of either amiodarone plus BB or and thyroid events and
than 20 consecutive d at anytime (patients sotalol vs BB alone (HR: 0.44; 95% symptomatic
Size: 412 patients who had received >10 d of amiodarone had CI: 0.280.68; p<0.001). Amiodarone bradycardia were more
to be taken off amiodarone for 10 d before plus BB significantly reduced the risk common among
randomization), a calculated creatinine of shock compared with BB alone patients randomized to
clearance of less than 30 mL/min (<0.50 (HR: 0.27; 95% CI: 0.140.52; amiodarone.
mL/s), symptomatic AF likely to require use p<0.001) and sotalol (HR: 0.43; 95%
of a class I or class III antiarrhythmic agent, CI: 0.220.85; p=0.02). There was a
absence of SHD, contraindications to trend for sotalol to reduce shocks
amiodarone or a -blocker, or NYHA class IV compared with BB alone (HR:
symptoms of HF. 0.61;95% CI, 0.371.01; p=0.055).
Intervention: amiodarone plus BB, sotalol The rates of study drug
alone discontinuation at 1y were 18.2% for
amiodarone, 23.5% for sotalol, and
Comparator: BB alone. 5.3% for BB alone.
Piccini et al. Aim: To evaluate Inclusion criteria: Studies in which patients 1 endpoint: SCD, CVD, all-cause Amiodarone reduces
2009 (154) the cumulative were randomized to amiodarone and mortality, and the incidences of drug the risk of SCD by 29%
19336434 evidence placebo or inactive control. Additional toxicities. and CVD by 18%,
regarding the safety inclusion criteria included: treatment for however, amiodarone
and efficacy of >30 d, follow-up >6 mo, and availability of Results: Amiodarone decreased the therapy is neutral with
amiodarone in all-cause mortality as an endpoint incidence of SCD (7.1 vs. 9.7%; OR: respect to all-cause
prevention of SCD 0.71; 95% CI: 0.610.84; p<0.001) mortality and was
Exclusion criteria: Studies and cardiovascular death (14.0% associated with a two-
Study type: Meta- of patients with shock-refractory VA, OHCA, vs.16.3%; OR: 0.82; 95% CI: 0.71 and five-fold increased
analysis of all RCT patients <18 y, randomization to 0.94, p=0.004). There was a 1.5% risk of pulmonary and
examining the use of amiodarone vs. a class Ic or class III AAD absolute risk reduction in all-cause thyroid toxicity.
amiodarone vs. (without a placebo or standard of care mortality which did not meet Authors suggested
placebo/control for arm). Studies of patients with ICDs were statistical significance (p=0.093). amiodarone as a viable
the prevention of excluded unless used on both arms. Amiodarone therapy increased the alternative in patients
SCD risk of pulmonary (2.9% vs. 1.5%; who are not eligible for
OR: 1.97; 95% CI: 1.273.04, or who do not have
Size: 15 trials, which p=0.002), and thyroid (3.6% vs. access to ICD therapy
randomized 8,522 0.4%; OR: 5.68; 95% CI: 2.9410.98, for the prevention of
patients p<0.001) toxicity. SCD.

111

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 26. Nonrandomized Trials, Observational Studies, and/or Registries of Secondary Prevention SCD in NICM (Section 7.2.1)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Raitt et al. 2001 Aim: To determine Inclusion criteria: 1 endpoint: Mortality Sustained VT without serious
(137) prognostic implications of Patients with stable VT symptoms or hemodynamic
11208684 stable VT that were not enrolled Results: The mortality in 440 compromise is associated with a
in AVID, were included patients with stable VT tended to high mortality rate and may be a
Study type: Observational, in a registry of patients be greater than that observed in marker for a substrate capable of
registry of patients with screened for the study. 1029 patients presenting with producing a more malignant
hemodynamically stable VT unstable VT (33.6% vs 27.6% at 3 arrhythmia
Exclusion criteria: y; RR:1.22; p=0.07). After
Size: The study population Patients who had an adjustment for baseline and
consisted of 440 patients arrhythmia within 5 d of treatment differences, the RR was
with stable VT and 1029 MI, cardiac surgery, or little changed (RR:1.25, p=0.06).
patients with unstable VT. Of coronary intervention
the 1029 patients with were excluded, as were
unstable VT, 330 had patients with NYHA class
therapy determined by IV HF or those who were
randomization in the AVID on a heart transplant
trial: 52% received an ICD, list, had a prior ICD
47% amiodarone, and 2% implant or attempted
sotalol. Therapy for the implant, or had a life
remaining 699 patients with expectancy of <1y.
unstable VT and the 440
patients with stable VT was
determined at the discretion
of the attending physician.
Ruwald et al. Aim: to evaluate (1) the Inclusion criteria: 1500 1 endpoint: Syncope was a 21 syncopal events (33%) were
2014 (196) effects of innovative ICD patients from 98 prespecified safety end point that classified as caused by VT or VF and
24201303 programming with either a hospital centers with a was adjudicated independently. 4 (6%) as caused by other or
high-rate cutoff VT zone or 1 prevention guideline Multivariable Cox models were unspecified arrhythmias, whereas a
delayed therapy on risk of indication to receive an used to identify risk factors total of 39 events (61%) were
syncope compared with ICD or CRT-D. associated with syncope and to classified as nonarrhythmogenic.
conventional programming; analyze subsequent risk of Syncope in HF patients (with a
(2) the independent Exclusion criteria: mortality. defibrillator) is primarily vasovagal,
prognostic factors associated Patients were excluded orthostatic, or otherwise

112

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
with syncope; and (3) the if they had experienced Results: Prognostic factors for all- nonarrhythmogenic in mechanism
association between AF within 1 mo before cause syncope included the and underscores the fact that the
syncope, the cause of implantation; if they presence of ischemic presence of heart disease (in this
syncope, and the risk of previously had been cardiomyopathy (HR: 2.48; 95% CI case, ischemic or nonischemic HF)
death in patients enrolled in implanted with a 1.424.34; p=0.002), previous VA does not dictate that syncope has a
MADIT-RIT pacemaker, ICD, or CRT- (HR: 2.99; 95% CI 1.187.59; cardiac cause
D; or if they had a recent p=0.021), LVEF 25% (HR: 1.65; Syncope in HF patients is related
Study type: Subgroup MI or revascularization 95% CI 0.982.77; p=0.059), and to an increased cardiovascular risk
analysis of MADIT-RIT. procedure (within 3 younger age (by 10 y; HR: 1.25; profile and is associated with an
mo). 95% CI1.001.52; p=0.046). increased risk of death regardless
Size: 64 of 1500 patients Syncope was associated with of its cause
(4.3%) had syncope increased risk of death regardless
of its cause (arrhythmogenic
syncope: HR: 4.51; 95% CI 1.39
14.64, p=0.012;
nonarrhythmogenic syncope: HR
2.97; 95% CI 1.078.28, p=0.038).
Middlekauff et Study type: Retrospective Inclusion criteria: 491 1 endpoint: Mortality Authors concluded that patients
al.1993 (3) cohort consecutive patients with advanced HF and syncope are
8417050 with advanced CHF Results: The actuarial incidence at especially high risk for sudden
Size: 491 patients with CHF, (NYHA functional class III of sudden death by 1 y was death regardless of the etiology of
of which 60 had a Hx of or IV), no Hx of CA and a significantly greater in patients syncope.
syncope; the condition had a mean LVEF of 0.20 with (45%) than in those without
cardiac origin in 29 (48%) 0.07. (12%, p<0.00001) syncope. In the
and was due to other causes Cox proportional hazards model,
in 31 (52%). syncope predicted sudden death
Exclusion criteria: N/A independent of AF, serum sodium,
cardiac index, angiotensin-
converting enzyme inhibition and
patient age. The actuarial risk of
sudden death by 1 y was similarly
high in patients with either cardiac
syncope or syncope from other
causes (49% vs. 39%, p=NS).
Knight et al.1999 Study type: Observational Inclusion criteria 1 endpoint: Mortality The authors conclude that the
(197) consecutive patients high incidence of appropriate ICD
10362200 Size: 14 patients who had a NICM, shocks and the association of

113

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
unexplained syncope Results: Seven of 14 patients recurrent syncope with VA support
and a negative (50%) in the Syncope Group the treatment of patients with
electrophysiology test received appropriate shocks for NICM unexplained syncope and a
and who underwent VA during a mean follow-up of negative electrophysiology test
defibrillator 2413 mo, compared with 8 of 19 with ICD.
implantation (Syncope patients (42%) in the Arrest Group
Group).19 consecutive during a mean follow-up of 4540
patients with a NICM mo (p=0.1).
and a CA who were
treated with a ICD
(Arrest Group) served as
a control group.

Exclusion criteria: N/A

Brilakis et al. Study type: Observational Inclusion criteria: Results: An EPS was done in 37 of The authors conclude that
2001 (198) Between 1990 and the 54 patients. In the 17 patients programmed ventricular
11816631 Size: 54 patients 1998, 54 (mean age who received an ICD, incidence of stimulation is not useful in risk
6711 y, 76% men) appropriate shocks at 1 and 3 y stratification of patients with IDCM
patients presented with was 47% and 74%, respectively, in and syncope and may delay
IDCM and syncope. the inducible sustained necessary ICD implantation.
monomorphic VT group, and 40%
Exclusion criteria: N/A and 40%, respectively, in the
group without inducible sustained
monomorphic VT (p=0.29, log-
rank test)
Fonarow et al. Study type: Observational Inclusion criteria: 147 Results: During a mean follow-up The authors conclude in patients
2000 (199) patients with Hx of of 22 mo, there were 31 deaths, with nonischemic cardiomyopathy
10760339 Size: 147 patients syncope and no prior Hx 18 sudden, in patients treated and syncope, therapy with an ICD is
of sustained VT or CA with conventional therapy, associated with a reduction in
were identified. whereas there were 2 deaths, sudden death and an improvement
Outcomes were none sudden, in patients treated in overall survival.
compared for the 25 with an ICD. An appropriate shock
patients managed with occurred in 40% of the ICD
an ICD and 122 patients patients. Actuarial survival at 2 y
managed with was 84.9% with ICD therapy and

114

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
conventional medical 66.9% with conventional therapy
therapy. (p=0.04).

Exclusion criteria: N/A

Olshansky et al. Study type: Subgroup Inclusion criteria: 1 endpoint: Outcomes, including Syncope was common in the
2008 (200) analysis of SCD-HeFT trial. Patients in the SCD-HeFT mortality, ICD discharges and SCD. SCD-HeFT population. Post-
18371559 trial who reported randomization syncope was
Size: 472 patients syncope prior of after Results: In SCD-HeFT, 162 (6%) associated with increased risk of
randomization. patients had syncope before all-cause mortality, cardiovascular
randomization, 356 (14%) had mortality, and SCD (despite
Exclusion criteria: N/A syncope after randomization randomization to an ICD). Those
(similar incidence in each patients randomized to an ICD,
randomized arm), and 46 (2%) had who had syncope, were more likely
syncope before and after to receive appropriate ICD shocks
randomization. In the ICD arm, than those without syncope; yet,
syncope, before and after did not protect patients against
randomization, was associated recurrent syncope and did not
with appropriate ICD discharges protect against the risk of death.
(HR: 1.75;95% CI: 1.102.80,
p=0.019 and HR: 2.91;95% CI:
1.894.47, p=0.001, respectively).
Post-randomization syncope
predicted total and cardiovascular
death (HR: 1.41; 95% CI: 1.13
1.76, p=0.002 and HR: 1.55; 95%
CI: 1.192.02, p=0.001,
respectively). The elevated
relative risk of mortality for
syncope vs. nonsyncope patients
did not vary significantly across
treatment arms (ICD, HR: 1.54;
95% CI: 1.042.27; amiodarone,
HR: 1.33; 95% CI: 0.911.93; and
placebo, HR: 1.39; 95% CI: 0.96
2.02, test for difference p=0.86).

115

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 27. RCTs Primary Prevention SCD in NICM (Section 7.2.2)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if any);
Author; Study Type; (# patients) / (Absolute Event Rates, Study Limitations;
Year Published Study Size (N) Study Comparator P values; OR or RR; & Adverse Events
(# patients) 95% CI)
CAT Aim: Multicenter Inclusion criteria: Intervention: ICD 1 endpoint: The 1 end Enrollment was terminated
Bnsch D et al. RCT of ICD vs. Recent onset of DCM (N=50) point of the trial was all- early because the interim
2002 (201) conventional (9 mo) and an EF 30% cause mortality at 1 y. analysis showed that the
11914254 Therapy in NIDCM and class II-III Comparator: overall1 y mortality rate for all
Conventional therapy Cumulative survival was patients was only 5.6%, well
Study type: RCT Exclusion criteria: CAD, (N=54) 92%, 86%, and 73% in the below the assumed value of
excessive alcohol ICD treatment group 30%.
Size: 104 patients intake, prior MI or vs. 93%, 80%, and 68% in Because the overall
myocarditis. the control group after 2, mortality rate was too low, the
4, and 6 y, respectively study was stopped for futility
(log rank p=0.554) after the pilot phase. Even if
1,348 patients had been
included, as initially planned,
the trial would have been
underpowered.

AMIOVIRT Aim: Multicenter Inclusion criteria: EF Intervention: ICD 1 endpoint: Total Trial terminated early for
Strickberger et RCT of ICD vs. 0.35, asymptomatic (N=51) Mortality futility in view of lower than
al. 2003 (202) amiodarone NSVT, NYHA class I to expected mortality.
12767651 Therapy in NIDCM III. Comparator: Survival at 1 y (90% vs. With the observed mortality
and NSVT Amiodarone (N=52) 96%) and 3 y (88% vs. rates, approximately 12,000
Exclusion criteria: 87%) was similar in the patients would have been
Study type: RCT Syncope, pregnancy, a amiodarone and ICD required to achieve a power of
contraindication to groups respectively 80%.
Size: 103 patients amiodarone or (p=0.8).
ICD or concomitant
therapy with a Class I
AAD

116

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
DEFINITE Aim: Multicenter Inclusion criteria: EF Intervention: ICD 1 endpoint: Total There were 3 sudden deaths
Kadish A, et al. RCT of ICD vs. 35%, and >10 PVCs/h (N=229) Mortality from arrhythmia in the ICD
2004 (203) standard medical or NSVT. group, as compared with 14
15152060 therapy in NIDCM Comparator: Fewer patients died in the deaths in the
and ambient VA Exclusion criteria: Conventional therapy ICD group than in the Control group (HR: 0.20; 95
NYHA class IV (N=229) Control group (28 vs. 40), % CI: 0.060.71; p=0.006)
Study type: RCT HF, familial but the difference in
cardiomyopathy survival was NS (p=0.08)
Size: 458 patients associated with sudden
death, acute
myocarditis or
congenital heart
disease.
SCD-HeFT Aim: Multicenter Inclusion criteria: Intervention: 1 endpoint: Amiodarone showed no
Bardy et al. RCT of ICD vs Ischemic or non Amiodarone (N=845) After a median follow-up benefit in survival
2005 (43) amiodarone vs. ischemic DCM, NYHA ICD therapy (N= 829) of 4 y, the mortality rate Non-ischemic DCM 48% of
15659722 optimal medical class II or III HF and was 22% in the ICD group, cohort.
therapy LVEF 35% Comparator: 28% in the amiodarone Similar benefit ischemic vs.
Optimal medical group, and 29% in the non-ischemic.
Study type: RCT Exclusion criteria: N/A therapy (N=847) control group. This
resulted in a 22% RR
Size: 2,521 reduction and a 7.2%
patients absolute risk reduction in
the all-cause mortality in
the ICD group as
compared with optimized
medical therapy alone
(p=0.007)
COMPANION Aim: Multicenter Inclusion criteria: Intervention: 1 endpoint: The 1 end A CRT pacemaker reduced
Bristow et al. RCT of CRT vs. CRT- 1,520 Ischemic or non CRT-D (N=595) point was a composite of the risk of the 2 end point of
2004 (204) D vs. optimized ischemic DCM, NYHA CRT Pacer (N=617) death or hospitalization death from any cause by 24%
15152059 medical therapy class III or IV, LVEF for any cause. (p=0.059), and a CRT
35% and QRS >120 Comparator: Optimal CRT-P decreased the risk pacemakerdefibrillator
Study type: RCT msec medical therapy of the 1 end point (HR: reduced the risk by 36%
(N=308) 0.81; p=0.014), as did CT- (p=0.003)
Size: 1,520 Exclusion criteria: N/A D (HR: 0.80; p=0.01). Non ischemic 44% of cohort
patients

117

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Desai et al. Aim: To determine Inclusion criteria: Intervention: ICD 1 endpoint: Five 1 Analysis of all 7 trials
2004 (195) whether ICD prospective RCTs of ICD prevention trials enrolling combined demonstrated a
15598919 therapy reduces all- or combined cardiac Comparator: 1854 patients with NICM statistically significant 31%
cause mortality in resynchronization Medical therapy were identified; pooled overall reduction in mortality
patients with NICM. therapy and analysis suggested a with ICD therapy (RR: 0.69;
defibrillator (CRT-D) vs significant reduction in 95% CI: 0.560.86; p=0.002).
Study type: meta- medical therapy total mortality among
analysis of RCTs enrolling at least some patients randomized to
individuals with NICM ICD or CRT-D vs medical
Size: 8 RCTs and reporting all-cause therapy (RR: 0.69; 95% CI:
enrolling a total of mortality as an 0.550.87; p=0.002).
2146 patients with outcome Mortality reduction
NICM were remained significant even
included. 7 trials after elimination of CRT-D
reported subgroup trials.
estimates for ICD
efficacy in NICM
DANISH Aim: To evaluate Inclusion criteria: Intervention: ICD 1 endpoint: Death from SCD (a 2 outcome) occurred
Kober L, et al. the benefit of Symptomatic patients (N=556) any cause. in 24 patients (4.3%) in the ICD
2016 (205) prophylactic ICDs in (NYHA class II or III, or group and in 46 patients
27571011 patients with NYHA class IV if CRT Comparator: Usual After a median follow-up (8.2%) in the control group
systolic HF that is was planned) with care for CHF (N=560) period of 67.6 mo, the 1 (HR: 0.50; 95% CI: 0.310.82;
not due to CAD nonischemic outcome had occurred in p=0.005)
systolic HF (LVEF 35%) 120 patients (21.6%) in 58% of patients received CRT
Study type: RCT and an increased level the ICD group and in 131 system, which could have
(>200 pg/mL) of N- patients (23.4%) in the influenced overall results.
Size: 1116 patients terminal probrain control group (HR: 0.87; Younger patients did show
natriuretic peptide (NT- 95% CI: 0.681.12; survival benefit.
proBNP). p=0.28).

Exclusion criteria:
Patients who had
permanent atrial
fibrillation with a
resting heart rate
higher than

118

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
100 beats per minute
or renal failure that was
being treated with
dialysis.

Data Supplement 28. Nonrandomized Trials, Observational Studies, and/or Registries of Primary Prevention of SCD in NICM (Section 7.2.2)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Marburg Aim: To determine Inclusion criteria: Men and 1 endpoint: During 5221 mo Non invasive tests such as signal-
Cardiomyopathy the women with IDC between 16 of follow-up, major arrhythmic averaged ECG, baroreflex
Study clinical value of and 70 y of age and LVEF <45% events were observed in 46 sensitivity, heart rate variability,
Grimm et al. 2003 potential noninvasive and a LV end-diastolic diameter patients (13%), including sudden and T-wave alternans did not seem
(206) arrhythmia risk >56 mm by echocardiography. cardiac death in 23 patients and to be helpful for arrhythmia risk
14623812 predictors in a large sustained VT or VF in another 23 stratification.
patient cohort with Exclusion criteria: CHF patients
IDC NYHA functional class IV; a Hx of
sustained VT or VF); an episode Results: On multivariate
of unexplained syncope within analysis, LVEF was the only
Study type: the previous 12 mo; class I or significant arrhythmia risk
Prospective class III AAD therapy that could predictor in patients with sinus
observational not be withdrawn for at least 5 rhythm, with a relative risk of 2.3
monocenter study drug half-lives; amiodarone per 10% decrease of LVEF (95%
therapy within the previous 6 CI: 1.53.3; p=0.0001). NSVT on
Size: 343 patients mo; pacemaker dependency; Holter was associated with a
CAD diagnosed by evidence of trend toward higher arrhythmia
any coronary artery stenosis risk (RR: 1.7; 95% CI: 0.93.3;
>50% by angiography; or a Hx of p=0.11), whereas BB therapy was
MI, systemic arterial associated with a trend toward
hypertension, active lower arrhythmia risk (RR: 0.6;
myocarditis, alcohol abuse, drug 95% CI: 0.31.2; p=0.13).
dependency, severe liver or
kidney disease, thyroid disease,
malignancies, or systemic
diseases.

119

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Goldberger et al. Aim: To estimate Inclusion criteria: 45 studies Results: Test sensitivities ranged Techniques incorporating
2014 (207) performance of 12 involving human subjects of the from 28.8% to 91.0%, functional parameters,
24445228 common risk following tests: baroreflex specificities from 36.2% to depolarization abnormalities,
stratification test as sensitivity, heart rate 87.1%, and odds ratios from 1.5 repolarization abnormalities, and
predictors of turbulence, heart rate to 6.7. Odds ratio was highest for arrhythmic markers provide only
arrhythmic events in variability, LV end-diastolic fragmented QRS and TWA (OR: modest risk stratification for
patients with DNICM dimension, LVEF, 6.73 and 4.66; 95% CI: 3.85 sudden cardiac death in patients
electrophysiologic study, NSVT, 11.76 and 2.558.53, with NICM.
Study type: meta- LBBB, signal-averaged respectively) and lowest for QRS At best, the OR for any 1
analysis of 12 electrocardiogram, fragmented duration (OR: 1.51; 95% CI: 1.13 predictor is generally in the range
commonly reported QRS, QRS-T angle, and T-wave 2.01). None of the autonomic of 2 to 4, precluding their
risk stratification alternans tests (heart rate variability, heart usefulness in isolation for
tests as predictors of rate turbulence, baroreflex individual patient decisions
arrhythmic events Exclusion criteria: N/A sensitivity) were significant
predictors of arrhythmic
Size: 45 studies outcomes.
enrolling 6,088
patients
Anselme et al. Aim: To evaluate a Inclusion criteria ICD implant at 1 endpoint: Malignant VA Life-threatening VAs are
2013 (208) strategy of any time during follow-up when common in patients with LMNA
23811080 prophylactic ICD in any of the following Results: ICD was implanted in 21 mutations and significant cardiac
LMNA mutation prespecified significant out of the 47 patients. Among conduction disorders, even if LVEF
carriers with conduction disorders was ICD recipients, no patient died is preserved
significant cardiac encountered: (1) requirement suddenly and 11 (52%) patients
conduction disorders for permanent ventricular required appropriate ICD therapy
pacing for bradycardia; (2) PR during a median follow-up of 62
Study type: interval >0.24 s and either mo. LVEF was 45% in 9 patients
Prospective single complete LBBB (LBBB) or NSVT; at the time of the event. Among
center observational (3) patients already implanted the 10 patients without
with a pacemaker at malignant VA, device memory
Size: 47 patients presentation to our center. recorded NSVT in 8 (80%). The
with LMNA presence of significant
mutations Exclusion criteria: N/A conduction disorders was the
only factor related to the
occurrence of malignant VA (HR:
5.20; 95% CI: 1.1423.53;
p=0.03).

120

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
van Rijsingen et al. Aim: The purpose of Inclusion criteria: Mutation 1 endpoint: First occurring Carriers of LMNA mutations with
2012 (209) this study was to carriers older than 15 y of age MVA. MVA were defined as a high risk of MVA can be identified
22281253 determine risk with a previously published appropriate ICD treatment, CPR, using these risk factors.
factors that predict pathogenic LMNA mutation or SCD Conduction disturbances were
malignant VA in with cardiac involvement and not a risk factor in this study.
Lamin A/C mutation persons with a newly identified Results: At median follow-up The 4 independent risk factors
carriers LMNA mutation with clinical or period of 43 mo (interquartile were NSVT, LVEF <45% at the first
family evidence of a range: 17101 mo), 48 (18%) clinical contact, male sex, and non-
Study type: laminopathy with possible persons experienced a first missense mutations (ins-del/
Multicenter, cardiac involvement. episode of MVA. Independent truncating or mutations affecting
retrospective risk factors for MVA were NSVT, splicing).
analysis Exclusion criteria: N/A LVEF <45% at the first clinical
contact, male sex, and non-
Size: 269 patients missense mutations (ins-
del/truncating or mutations
affecting splicing). MVA occurred
only in persons with at least 2 of
these risk factors. There was a
cumulative risk for MVA per
additional risk factor.
Pasotti et al. 2008 Aim: The aim of this Inclusion criteria: 27 1 endpoint: Events were death Authors concluded that dilated
(210) study was to analyze consecutive families in which from any cause, death from HF, cardiomyopathies caused by LMNA
18926329 the long-term follow- LMNA gene defects were heart transplantation, and SCD, gene defects are highly penetrant,
up of dilated identified in the probands, all including appropriate ICD adult onset, malignant diseases
cardiolaminopathies sharing the DCM phenotype. Of interventions characterized by a high rate HF and
in patients with the 164 family members, 94 had life-threatening arrhythmias.
LAMIN gene LMNA gene mutations Results: Neither AVB nor pacemaker
mutations 60 of 94 (64%) were implantation turned out to be
Exclusion criteria: N/A phenotypically affected whereas predictors of events.
Study type: 34 were only genotypically NYHA class III to IV and highly
Retrospective affected. dynamic
observational Of the 60 patients, 40 had DCM Competitive sports for 10 y were
longitudinal study with AVB, 12 had DCM with independent predictors of total
VT/VF, 6 had DCM with AVB and events.
Size: 94 patients EDMD2, and 2 had AVB plus
EDMD2.

121

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
During a median of 57 mo there
were 49 events in 43 DCM
patients.
The events were related to HF
(15 heart transplants, 1 death
from end-stage HF) and VA (15
SCDs and 12 appropriate ICD
interventions).
van Berlo et al. Aim: To evaluate Inclusion criteria: 21 1 endpoint: Arrhythmias and Authors conclude that carriers of
2005 (211) common clinical publications between March sudden death lamin A/C mutations carry a high
15551023 characteristics of 1999 and March 2002 reporting risk of sudden death.
patients with lamin lamin A/C gene mutations Results: Presence of pacemaker did not
A/C gene mutations Cardiac dysrhythmias were protect against sudden death.
that cause either Exclusion criteria: Patients reported in 92% of patients after
isolated DCM or DCM with familial partial the age of 30 y; HF was reported
in association with lipodystrophy, progeria, axonal in 64% after the age of 50.
skeletal muscular neuropathy and mandibuloacral 76 of the reported 299 patients
dystrophy. dysplasia caused by mutations (25%) died at a mean age of 46 y.
in the lamin A/C gene were Sudden death was the most
Study type: Meta- excluded frequently reported mode of
analysis (pooled death (46%) in both the cardiac
data) and the neuromuscular
phenotype.
Size: 299 carriers of
lamin A/C mutations
Piccini et al. 2009 Aim: To evaluate the Inclusion criteria: Studies in 1 endpoint: SCD, CVD, all-cause Amiodarone reduces the risk of
(154) cumulative evidence which patients were mortality, and the incidences of SCD by 29% and CVD by 18%,
19336434 regarding the safety randomized to amiodarone and drug toxicities. however, amiodarone therapy is
and efficacy of placebo or inactive control. neutral with respect to all-cause
amiodarone in Additional Results: Amiodarone decreased mortality and was associated with
prevention of SCD inclusion criteria included: the incidence of SCD [7.1 vs. a 2- and 5-fold increased risk of
treatment for >30 d, follow-up 9.7%; OR: 0.71; 95% CI 0.61 pulmonary and thyroid toxicity.
Study type: Meta- >6 mo, and availability of all- 0.84; p<0.001] and Authors suggested amiodarone
analysis of all RCT cause mortality as an endpoint cardiovascular death (CVD) as a viable alternative in patients
examining the use of [14.0% vs.16.3%; OR: 0.82; 95% who are not eligible for or who do
amiodarone vs. Exclusion criteria: Studies CI 0.710.94, p=0.004]. There not have access to ICD therapy for
placebo/control for was a 1.5% absolute risk the prevention of SCD.

122

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
the prevention of of patients with shock- reduction in all-cause mortality
SCD refractory VA, OHCA, patients which did not meet statistical
<18 y, randomization to significance (p=0.093).
Size: 15 trials, which amiodarone vs. a class Ic or Amiodarone therapy increased
randomized 8,522 class III AAD (without a placebo the risk of pulmonary [2.9% vs.
patients or standard of care arm). 1.5%; OR: 1.97; 95% CI 1.27
Studies of patients with ICDs 3.04, p=0.002], and thyroid [3.6%
were excluded unless used on vs. 0.4%; OR: 5.68; 95% CI 2.94
both arms. 10.98, p<0.001] toxicity.

WEARIT-II Study type: Inclusion criteria: All patients 1 endpoint: There was a total of 120
Kutyifa et al. 2015 Observational with LifeVest offered sustained ventricular
(212) patients with LVEF and a high Results: tachyarrhythmias in 41 patients, of
26316618 Size: 2000 risk for SCD after MI, following 805 patients (40%) had ischemic whom 54% received appropriate
coronary revascularization, with cardiomyopathy, 927 patients WCD shock. Only 10 patients
a new-onset dilated NICM, with (46%) had nonischemic (0.5%) received inappropriate WCD
high risk for SCD until cardiomyopathy, and 268 (14%) therapy.
stabilization, or with inherited patients were diagnosed with The rate of sustained ventricular
or congenital heart disease congenital or inherited heart tachyarrhythmias by 3 mo was 3%
Exclusion criteria: refused disease among patients with ischemic
consent The median age was 62 y; the cardiomyopathy and
median LVEF was 25%. The congenital/inherited heart disease,
median WCD wear time was 90 and 1% among nonischemic
d, with median daily use of 22.5 patients (p=0.02).
h. 90 sustained VT events in 22
patients were withheld from
therapy, whereas 30 events in 22
patients required WCD shock
therapy owing to hemodynamic
instability (corresponding to 5
events per 100 patient y).
All patients who required shock
delivery had their VT/VF episodes
successfully terminated with the
first shock.
10 patients (0.5%, 2 per 100
patient-y) had inappropriate WCD

123

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
therapy during the follow-up
because of ECG artifacts.
Inappropriate shocks did not
induce VT or VF.
Singh et al. 2015 Study type: Inclusion criteria: All 1 endpoint: Appropriate WCD Single center study
(213) observational single consecutive patients prescribed therapy
26670060 center a WCD between June 1, 2004
and May 30, 2015 at the Results: During 56.7 patient-y, 0
Size: 691 (254 new hospitals comprising the NICM patients received an
NICM and 271 new University of Pittsburgh Medical appropriate WCD shock
ICM Center to which access to
clinical data was available. During 46.7 patient-y, 6 (2.2%)
ischemic
Exclusion criteria: cardiomyopathypatients received
Patients with an explanted ICD an appropriate shock; 5
awaiting reimplantation, prior survived the episode, and 4
cardiac arrest unrelated to AMI, survived to hospital discharge
or elevated risk of SCD for
reasons other than ICM or
NICM.
Uyei et al. 2014 Study type: N/A 1 endpoint: N/A The quality of evidence was low
(214) Systematic review to very low quality, such that our
24893969 Results: It appears that confidence in the reported
wearable defibrillator use estimates is weak.
Size: compared with no
defibrillator use reduces the
chance of VT/VF associated
deaths by an absolute risk
reduction of approximately 1%,
achieved by averting
approximately 4/5th of all VT/VF
associated deaths.
Al-Khatib et al. Study type: meta- Inclusion criteria: 1 1 endpoint: all-cause mortality 1 prevention ICDs are efficacious
JAMA Cardiology analysis of RCTs prevention ICDs in patients with at reducing all-cause mortality in
2017 (215) NICM Results: patients with NICM
28355432 Size: N=1,874 Pooling data with fixed and RE
Exclusion criteria: models from these 4 studies

124

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
CRT showed a significant reduction in
Antiarrhythmic medication arm all-cause mortality with an ICD
(HR: 0.75; 95% CI 0.61-0.93, p=
0.008; p for
heterogeneity=0.873)

Data Supplement 29. RCTs and Nonrandomized Trials, Observational Studies, and/or Registries of Treatment of Recurrent VA in Patients
With NICM (Section 7.2.3)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
OPTIC Study Aim: To determine Inclusion criteria: Patients 1 endpoint: ICD shock for any Amiodarone plus BB is effective
Connolly et al. whether amiodarone were eligible if they had reason. for preventing these shocks and is
2006 (159) plus BB or sotalol are received an ICD within 21 d more effective than sotalol but has
16403928 better than BB alone for inducible or Results: Shocks occurred in 41 an increased risk of drug-related
for prevention of ICD spontaneously occurring VT patients (38.5%) assigned to BB adverse effects
shocks. or VF. alone, 26 (24.3%) assigned to Adverse pulmonary and thyroid
sotalol, and 12 (10.3%) assigned to events and symptomatic
Study type: Exclusion criteria: Patients amiodarone plus BB. A reduction in bradycardia were more common
multicenter RCT were excluded if they had the risk of shock was observed with among patients randomized to
LQTS, corrected QT interval of use of either amiodarone plus BB amiodarone.
Size: 412 patients more than 450 millisec, were or sotalol vs BB alone (HR: 0.44;
receiving a class I or class III 95% CI: 0.280.68; p<0.001).
antiarrhythmic agent, had Amiodarone plus BB significantly
received amiodarone or reduced the risk of shock
sotalol for more than 20 compared with BB alone (HR: 0.27;
consecutive days at anytime 95% CI: 0.140.52; p<0.001) and
(patients who had received sotalol (HR: 0.43; 95% CI: 0.22
>10 d of amiodarone had to 0.85; p=0.02). There was a trend
be taken off amiodarone for for sotalol to reduce shocks
10d before randomization), a compared with BB alone (HR:
calculated creatinine 0.61;95% CI, 0.371.01; p=0.055).
clearance of less than 30 The rates of study drug
mL/min (<0.50 mL/s), discontinuation at 1y were 18.2%
symptomatic AF likely to for amiodarone, 23.5% for sotalol,
require use of a class I or and 5.3% for BB alone.

125

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
class III antiarrhythmic agent,
absence of SHD,
contraindications to
amiodarone or a -blocker, or
NYHA class IV symptoms of
HF.
Intervention: amiodarone
plus BB, sotalol alone

Comparator: BB alone.
International Aim: to determine the Inclusion criteria: SHD with 1 endpoint: Procedural complications
VT Collaborative association of VT Ischemic and Non-Ischemic Freedom from VT recurrence, occurred in 6%, including 2 deaths
Group Study recurrence after cardiomyopathies with Heart Transplant, or death was (0.1%), hemopericardium in 1.7%,
Tung R 2015 ablation and survival in monomorphic VT and 70% at 1 y follow-up. and vascular access complications in
(178) scar related VT myocardial scar by VT recurred in 55% of patients who 1.6%
electroanatomic mapping died vs. 22% of patients who
Study type: survived.
Multicenter Exclusion criteria: absence of Transplant free survival was 90%
observational scar on electroanatomical for patients without VT recurrence
mapping and 71% for those with VT
Size: 2061 recurrence (HR 6.9; 95% CI 5.39.0,
Intervention: Catheter p<0.001).
ablation, either endocardial
or epicardial, guided by EAM.
End point of ablation with
elimination of all induced VTs

HELP-VT Aim: Inclusion criteria: 1 endpoint: At 1y follow-up, VT Complications


Dinov 2014 To determine the Patients with SHD referred free survival was 57% for ischemic Complications occurred in 11.1% of
(175) outcome of VT for catheter ablation of VT cardiomyopathyand 40.5% for NICM and 11.1% of ischemic
24211823 catheter ablation in with either NICM (N=63) or NICM patients (HR: 1.62; 95% CI: cardiomyopathypatients, including
patients with NICM to ischemic 1.122.34, p=0.01). ischemic death in 4.8% of NICM and 3.7% of
those with Ischemic cardiomyopathy(N=164) cardiomyopathyrequired epicardial ischemic cardiomyopathy
Cardiomyopathy (ICM) Exclusion criteria: ablation in only 2 of 164 (1.2%)
Study type: Failure of informed consent whereas NICM required epicardial
Prospective, non- Intervention: ablation in 30.8% (p=0.0001).
randomized

126

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 227 patients Catheter ablation for
patients with NICM
Comparator:
Catheter ablation in patients
with ischemic
cardiomyopathy

Data Supplement 30. Nonrandomized Trials, Observational Studies, and/or Registries of Arrhythmogenic Right Ventricular Cardiomyopathy
(Section 7.3)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Quarta G, et al. Study type: Inclusion criteria: 1 endpoint: Familial evaluation for ARVC; >50% probands died suddenly
Circ 2011 (216) national cohort 100 families with followup 3.41.6 y. Deceased proband in 51 Desmosomal gene complexity in
21606390 ARVC evaluated families 10% of relatives, assoc with 5-fold
Size: 255 2003-2009 increased risk of disease expression
Results: in 88% of deceased: dx of ARVC made
first degree: 210 at autopsy
second degree: 45 SCD most common in young: 31% died
between 14-20 y
Exclusion criteria: Definite or probable gene mutations; 58% of
N/A families, 73% of living probands
42% of first degree relatives had disease
expression
62% of gene carriers had phenotypic
expression
Progressive disease expression beyond age 40
in 50%
Kapplinger JD Study type: Multi- Inclusion criteria: 1 endpoint: Determine prevalence of Radical mutations are high-
JACC 2011 (217) center Netherlands, ARVC patients and background noise in ARVC genetic testing probablility ARVC associated
21636032 retrospective 427 unrelated mutations
healthy controls Results: Mutations present in 58% of ARVC R Missense mutation should be
Size: 93 probands and 16% of controls interpreted in context of race,
and 427 controls Radical mutations: 43% of ARVC, vs 0.5% ethnicity, mutation location,
controls sequence conservation; more likely
127

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Tested for PKP2, Missense mutations: 21% of ARVC, 16% of positive if Caucasian, within DSP and
DSP, DSG2, DSC2, controls DSG2 hotspot, and conserved in
TEME43 PKP2 and DSG2 residue
R Background mutation rate = 16%
Added data from 82 (vs 5% for LQT1-3)
patients in ARVD/C
Registry in USA

Exclusion criteria:
N/A
Bhonsale A, et al. Study type: Inclusion criteria: 1 endpoint: Risk stratification in ARVC ARVC desmosomal mutation
CAE 2013 (218) ARVC patients with genotype positive: sustained VT, SCD/ADA, carriers risk stratification:
23671136 Size: 215 positive genotype: appropriate ICD shock High risk: ECG 3 T wave
desmosomal Mean followup 7 y inversions, Holter, proband status
mutation carriers Increasing PVCs on holter c/w
PKP2 85% Results: 40% ACE arrhythmic events, > 760 PVC
53% males, mean ECG: high risk 3 inverted precordial T waves; Benign ECG conferred low
age 32 18 y intermediate risk = T wave inversion in leads arrhythmic risk
Presentation VT/VF V1, V2 + late depol; low risk = 02 T wave
23% inversion without depol changes
PVC count on holter higher in arrhythmic
Exclusion criteria: outcomes, p<0.0001
N/A Event free survival lowest among probands
p<0.001, and symptomatic patients p<0.001
Incremental risk: Proband, HR: 7.7; 3 T wave
inversions, HR: 4.2; male gender, HR: 1.8
Marcus FI, et al. Review paper for physicians summarizing ARVC: aut dominant, Desmosomes: cardiac, Proband may not benefit from
JACC 2013 (219) genetics of ARVC skin, hair gene testing, does not alter therapy.
23500315 30-50% of patients with ARVC have abnormal Patients with >1 gene abnormality
5 genes: gene, range 26-58%, highest in clinical familial may have more severe course;
Plakophilin- 2 73-78% disease. 20-30% family Hx sudden death earlier ICD.
Desmoglein -2 10-13% Benefits genetic testing ARVC:
Desmocollin-2 4-6% Negative genetic tesing no disease, as >50% understand cause of disease, identify
Desmoplakin 3-8% gene negative to date. family members at risk, family
Junctional 1-4% Abnormal gene = risk, but not disease; planning, limited prognostic
plakoglobin modified by additional gene modifiers, virus, information.
athletics

128

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Cost ~$5400 PKP2 may require a second mutation to cause For gene carriers: Recommend
disease. The second mutation may not be cardiac eval beginning at 10-12 y:
tested in relatives, leading to false negative. ECG, SAECG, echo, holter, CMR
~48% of patients with ARVC have at least 2 Evaluate q 2 y between 10-20 y;
different mutations; these patients have more then every 5 y, may stop at age 50-
severe disease. 60 y.
Truly abnormal gene should not be present in
>1:400 controls;
However, 1:200 Finnish have desmosomal
mutation of ARVC; 6% of Asians carry PKP2
mutations.
the interpretation of genetic results for ARVC
is not an exact science and is more complex
than for other heart disorders caused by only
a single gene and for which most patients will
have an abnormal gene identified.

Bhonsale A et al. Study type: Inclusion criteria: 1 endpoint: Impact of genotype on clinical Among ARVC patients with known
Eur Heart J 2015 Retrospective Genotype positive course in ARVC mutation carriers. Mean genotype: specific genotype affects
(220) multicenter, Dutch, desmosomal and followup 67 y. clinical course and disease
25616645 US non-desmosomal expression.
mutations in ARVC. Results: Presentation with SCD were younger Gene specific variation in SCD, LV
Size: 577 PKP2 80% (median 23 y) than those presenting with VT dysfunction, HF.
(36 y) (p<0.001). Males worse outcome: more likely
Males 55%, mean Death 2%, transplant 2%; Sustained VT/VF to be probands, symptomatic earlier
age 3517 y. 541 30%, LVEF < 55 14%; CHF 5%. and more severe arrhythmic
presenting alive: Compound mutations: earlier onset of expression.
Presentation SCD= symptoms, higher incidence VT/VF. PKP2 least Phenotypic variabilitymodifier
6% ventricular dysfunction, 9%; genes/environmental influences.
41% probands.
Desmoplakin (DSP) mutations had more
Exclusion criteria: ventricular dysfunction/HF than PKP2 carriers:
non-genotyped 40% ventricular dysfunction; more likely to
ARVD present with SCD (11% of SCD)

Male gender higher arrhythmic outcome, 53%


vs 29%

129

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Rigato I et al. Circ Study type: Inclusion criteria: 1 endpoint: ARVC gene carriers risk of Multiple DS gene mutation status
CV Genetics 2013 Prospective Desmosomal gene arrhythmic outcome was powerful predictor for major
(221) Observational mutations carriers arrhythmic events.
24070718 Desmoplakin 39%, Results: Median observation 39 y (22-52)
Size: 134 plakophilin 2 34%, 16% major arrhythmic events.
desmoglein 2 26%, Independent predictors:
desmocolliln 2 1% Multiple desmosomal gene mutations HR:
16% complex 3.71; 95 CI:1.548.92, p=0.003.
genotype: Male gender HR: 2.76; 95% CI: 1.196.41,
compound or p=0.02.
dignenic
heterozygosity

Exclusion criteria:
N/A
Groeneweg JA et Study type: Inclusion criteria: 1 endpoint: outcomes of ARVC patients ARVC: 10% death/heart
al. Circ CV Genetics retrospective ARVC patients median followup 7 y transplantation during median
2015 (222) multicenter, Europe Probands 44%, followup 7y.
25820315 and USA family members Results: Sustained VT developed in 72% of Probands: Mutations altered age of
56%. probands. disease expression but not
Size: 1001 Probands: 416/439 Probands with positive mutations presented outcomes.
presented alive (5% at younger age. Family members: mutation
presented SCD). Mortality 6%, transplantation 4%, not carriers had more VA and increased
different based on mutation status in cardiac mortality.
Overall 63% probands.
mutation positive: Family members: 1/3 developed ARVC.
PKP2 46%. Sustained VT 8%, cardiac mortality 2%.
Family members:
73% mutation Mutations in family members modified
carriers. course: 8x increase in VT, increased cardiac
mortality.
Exclusion criteria: ICD improved survival in index patients: SCD
N/A 0.6% vs 16% without ICD.

130

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
te Riele AS, et al. Study type: Inclusion criteria: 1 endpoint: ARVC first degree relatives: risk ARVC first degree relatives with
EHJ 2016 (223) Multicenter First degree of ARVC dx and outcomes increased likelihood of dx:
26314686 retrospective relatives of ARVC Mean followup 6.73.7 y symptoms, sibling, pathogenic
proband mutation, female gender.
Size: 274 46% male, age Results: 35% developed ARVC Malignant family Hx was not
3619 y Risk of ARVC dx: sibling, HR: 3.11; p<0 .001, associated with arrhythmic events
symptoms, p<0.001, pathogenic mutation
Exclusion criteria: p<0.001, female, p=0.01.
N/A 8% developed sustained VA: neither
relatedness to proband nor malignant family
Hx were predictive of arrhythmic events.
Kamath GS, et al., Study type: Inclusion criteria: 1 endpoint: SAECG abnormalities in ARVC SAECG: using 1/3 criteria increased
HR 2011 (224) retrospective single ARVC probands Abnormal: fQRSD 114 ms, LASD >38 ms, sensitivity and maintained specificity
20933608 center compared with 103 RMS-40 <20 V SAECG correlated with disease
controls severity on CMR, but not VT
Size: 87 Results:
Mean age 37 y, 54% SAECG sensitivity/specificity: 1-criteria 69%/
male 92%; 2-criteria 47%/95%; 3-criteria 33%/100%

Exclusion criteria:
N/A
Marcus FI, et al., Study type: Single Inclusion criteria: 1 endpoint: right ventricular abnormalities in Characterize RV pathology in LBBB
Circ 1982 (225) center 22 adults with ARVC VT
7053899 recurrent VT w/ Consider dx in patients with VT of
Size: 22 LBBB 21/22 Results: inverted T waves right precordium, unknown cause, particularly if LBBB
Mean age 39 y, cardiac enlargement, delayed ventricular pattern
Males 2.7:1 potentials
RV dysplasia inferior, apical or
Exclusion criteria: diaphragmatic-diagnosed with angiography. 1
N/A death.
Corrado D et al. Study type: Inclusion criteria: 1 endpoint: ARVC clinic-pathologic LV involvement in 76% of ARVC:
JACC 1997 (226) retrospective Pathologic dx of manifestations age dependent,
9362410 multicenter ARVC at autopsy or more severe cardiomegaly
heart transplant Results: 80% died suddenly: 47% of SCD died More CHF
Size: 42 Mean age 29.618 y during exertion Prior syncope in 26%
(965 y) SCD first symptom in 35%. SCD exercise related in 47%
CHF 24%

131

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: Syncope 26%
N/A Exercise related in 64%
LV fibrofatty involvement 76%
Isolated RV involvement 24%
Link MS ert al. Study type: Inclusion criteria: 1 endpoint: Sustained VA in ARVC during ARVC predictors of VT: sustained
JACC 2014 (227) Prospective multi- ARVC patients followup 3.31.7 y VT prior to ICD, inferior T wave
25011714 center enrolled in registry inversion, younger age at enrollment
North American Results: 44% (48 patients) had 502 episodes 48% received ICD therapy
ARVC Registry 79% (108 patients) of sustained VT: 97% monomorphic VT. Recommend programming ATP for
received ICDs Inapprop shocks 17%. termination of VT: successful 92%
Size: 137 Independent predictors sust VT: prior Syncope, family Hx SCD did not
Mean age spontaneous VT, inferior T wave inversion. predict ICD therapy
enrollment 4014 y. Independent predictor life threatening VT
Prior symptoms, (rate 240bpm or VF): younger age at
sustained VT or CA enrollment.
41% ATP successfully terminated 92% of VT
Patients without ICD implantation: no SCD or
SVT -followup 2.4 y
Exclusion criteria:
N/A
Corrado D et al. International Task Force No competitive or endurance sports; AADs as ICD implantation:
Circ 2015 (228) adjunct in patients w frequent AICD shocks; Hemodynamically unstable sust VT,
26216213 Treatment of ARVC: International Task BB for patients with recurrent VT, appropriate or VF; severe systolic dysfunction RV
Force Recommendations ICD rx, or ICD therapy for SVT; epicardial or LVEF 35%;
ablation for patients who fail endocardial Hemodynamically stable sustained
approach; ICD for patients with hemo VT; unexplained syncope; mod vent
unstable sustained VT/ VF. dysfunction RV EF= 36-40% or LVEF=
36-45%; or NSVT
EPS for suspected ARVC; restrict athletics to Minor risk factors
low intensity; BB for all ARVC patients Prophylactic ICD in asymptomatic
irrespective of arrhythmias; cath ablation for patients with no risk factors of
recurrent VT fail meds other than amio. healthy gene carriers.

Vstim for risk stratification asymptomatic;


endocardial voltage mapping; restrict comp
sports in phenotype neg patients; cath
ablation without ICD for selected patients

132

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
with drug refractory hemo stable single
morphology VT.

No BB for healthy gene carriers; cath ablation


as alternative to ICD for prevention of SCD.
Corrado D et al. Study type: Inclusion criteria: 1 endpoint: ARVC appropriate ICD shocks 48% approp ICD shocks
Circ 2003 (229) multicenter ARVC patients with Mean followup 39 mo Predictors: ACA, unstable VT,
14638546 retrospective ICD younger age, lower LVEF
Mean age 40 y Results: Approp shocks 48%, comps 14%, PES not predictive of approp shock
Size: 132 70% males inapprop shocks 16% Syncope not statistically important
ICD indication: ACA 84% underwent PES: 69% inducible sust VT: as risk factor in multivariable
10%, sustained VT neither sensitive nor specific: 51% no appropr analysis.
62%, syncope 16%; shock, 54% of non-inducible had approp rx 4 patients implanted due to family
nonsust VT 9%; Syncope: 21 patients: none died, one Hx SCD: no approp shocks
family Hx 3% underwent OHT; 38% approp shocks;
multivariate analysis p=0.07 for approp shock
83% on AA drugs Independent predictors of VF: ACA, VT with
prior to ICD hemodynamic compromise, younger age, LV
involvement
Exclusion criteria:
N/A
Piccini JP et al. Study type: single Inclusion criteria: 1 endpoint: ARVC clinical + EP Multivariate predictor approp
Heart Rhythm 2005 center retrospective Patients with characteristics that predict appropriate ICD shock: sustained VT/VF, OR:11.4;
(230) definite or probable shocks. p=0.015;
16253908 Size: 67 ARVC with ICDs Mean followup 4.42.9 y NSVT, OR: 6.29, p=0.051
Mean age 3614 y; EPS did not predict ICD shocks in
52% male Results: Appropriate shocks in 94% of 2 patients with 1 prevention ICD
1 prevention 42%, prevention, 39% of 1 prevention (p=0.001), Further research to identify low
2 58% overall 66% risk patients who do not need ICD
Sustained VT: 52%, approp shocks: Definite ARVC: 73%; placement
syncope 36%, ACA probable:33% Syncope not statistically significant
58/5 Overall 21% received shock for life
threatening VT/VF >240 bpm; no difference in
1 or 2 prevention patients
Exclusion criteria: EPS did not predict ICD approp use in patients
N/A with 1 prevention
All patients with VF had inducible VT/VF

133

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Syncope: 43% approp shocks, 22% no rx,
p=0.08

Bhonsale A et al. Study type: Inclusion criteria: 1 endpoint: Incidence and predictors of 48% ARVC patients undergoing 1
JACC 2011 (231) Retrospective single Definite or probable appropriate ICD shocks for ARVC undergoing prevention ICD received appropr
21939834 center ARVC with ICD ICD for 1 prevention shocks
implantation for 1 Mean followup 4.73.4 y. Approp shocks: proband, inducible at
Size: 84 prevention EPS, clinical nonsust VT, PVCs
63 patients Results: 48% approp ICD shocks. >1000/24 hrs
genotyped: 43% + Predictors: Multivariable analysis: Positive VT
desmosomal inducibility at PES, HR: 4.5; 95% CI: 1.415, Syncope NS predictor, HR: 0.91
mutations p=0.013), clinical nonsust VT, HR:10.5; 95% CI:
2.446.2, p=0.002); PVCs >1000/24 h, HR: Non-inducible: 1/20 appropr ICD
76% symptomatic, 3.48; proband, HR:1.62. shock
63% >1000 PVCs
on holter Syncope: approp shocks 9%/y. 25% approp
shocks, vs 30% no approp shocks
Syncope: 27% Recent syncope <6 mo: 63% appropr shocks
Exclusion criteria: vs 20% remote, p=0.046
N/A
Dalal D et al. JACC Study type: Inclusion criteria: 1 endpoint: Efficacy of ablation for ARVC. High rate of recurrent VT after
2007 (232) retrospective single ARVC patients Mean followup 32 mo. ablation for ARVC
17662396 center undergoing ablation diffuse cardiomyopathy with
at Hopkins. Results: 48 procedures. 46% eliminated all evolving electrical substrate
Size: 24 inducible VT
Mean age 369 y, Recurrence: overall 85%. One procedural
46% males death 4%. VT recurrence free survival: 50% at
5 mos, 25% at 14 mo. Did not vary by
Exclusion criteria: procedural success, mapping, repeat
N/A procedures.

134

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Garcia FC et al. Study type: Inclusion criteria: 1 endpoint: Endocardial vs epicardial Epicardial ablation in ARVC after
Circ 2009 (233) retrospective single ARVC patients ablation in ARVC failed endocardial ablation results in
19620503 center undergoing VT control
epicardial ablation Results: 27 VTs in 13 patients
Size: 13 after failed 85% epi ablation opposite endocardial
endocardial ablation sites
ablation VT 77% no VT with 1813 mo followup

Exclusion criteria:
N/A
Philips B et al. Circ Study type: Inclusion criteria: 1 endpoint: ARVC Efficacy of epicardial Epicardial ablation of VT in ARVC
AE 2012 (234) Retrospective ARVC patients ablation of VT. associated with high recurrence rate,
22492430 multicenter undergoing ablation but reduces VT burden.
1992-2011 at 80 Results: 175 ablations in 87 patients: 53% Majority of VT circuits were
Size: 87 centers. repeat procedures. epicardial.
Mean age 3311 y, 27% recurrent VT; VT reduction
53% male Freedom from VT at 1, 5, 10y: 47%, 21%, 15%.
50% failed Epicardial ablation: freedom from VT at 1, 5 y:
endocardial 64%, 45%
ablation Burden of VT reduced irrespective of ablation
strategy: p<0.001
Exclusion criteria: Complications: 2.3% major: death; delayed
N/A MI/occlusion RCA. Related to pericardial
access.
Bai R, et al. CAE Study type: Inclusion criteria: 1 endpoint: Comparison of outcomes for Combined endocardial-epicardial
2011 (235) Multicenter Consecutive ARVC ARVC ablation, endocardial vs endo- ablation approach in ARVC achieves
21665983 prospective patients undergoing epicardial: non-inducibility of VT with isuprel. longer term freedom from VA or
ablation Followup 3 y shocks.
Size: 49 All sust Patients with frequent PVCs more
monomorphic VT; Results: Freedom from VA or ICD therapies: likely to have recurrences
all with AICDs Endocardial: 52%, endo-epi 85%, p=0.029

Exclusion criteria:
N/A
Berruezo A et al. Study type: Inclusion criteria: 1 endpoint: ARVC patients: recurrence of VT ARVC combined endo + epi
Circ AE 2012 (236) retrospective single ARVC patients after ablation endo + epicardial ablation reveals wider substrate,
22205683 center undergoing endo + with good short/mid-term success

135

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
epicardial ablation Results: ablation eliminated all clinical and
Size: 11 of VT induced VT
64% continued on sotalol
Exclusion criteria: 9% VT recurrence with median 11 mo
N/A followup
Philips B Heart Study type: Inclusion criteria: 1 endpoint: Safety and efficacy of epicardial Epicardial ablation for VT in ARVC
Rhythm 2015(237) retrospective single ARVC undergoing ablation at tertiary center for ARVC safe in tertiary center
25530221 center epicardial ablation Freedom from VT 70% at 2 y.
at tertiary center Results: VT circuits: 69% on epicardial Reduces VT burden
Size: 30 surface, most sub-tricuspid. VT recurrence:
Exclusion criteria: 27%.
N/A Reduced VT burden (p<0.001)
VT free survival at 1,2 y: 76%, 70%
Complications: 3.3%, pericarditis. Fluoro 82
min (40-135)
Santangeli P et al. Study type: Inclusion criteria: 1 endpoint: ARVC ablation outcomes, ARVC VT ablation outcomes good;
Circ AE 2015 (238) Retrospective single ARVC patients followup 5644 mos most have VT control
26546346 center undergoing ablation Epicardial ablation if failed endocardial
Endo + epi: 63% ablation
Size: 62
Exclusion criteria: Results: VT recurrence: 29%; VT free survival
N/A 71%
64% on BB or no rx
James CA et al. Study type: Single Inclusion criteria: 1 endpoint: ARVC exercise and VT/VF Endurance and frequent exercise
JACC 2013 (239) center retrospective ARVC patients increase the risk of VT/VF, HF in
23871885 interviewed about Results: Endurance athletes developed ARVC patients.
Size: 87 exercise from 10 y symptoms at younger age (3013 y) vs 40 y,
of age. p=0.05;
Mean age 4418 y Increasing exercise
Lower lifetime survival free of VT/VF p=0.013
Exclusion criteria:
N/A
Sawant AC et al. Study type: single Inclusion criteria: 1 endpoint: ARVC: exercise and impact on Gene-elusive non-familial ARVC is
JAHA 2014 (240) center retrospective ARVC patients desmosomal and gene-elusive patients assoc with very high intensity
25516436 interviewed re exercise
Size: 82 exercise Recommend exercise restriction

136

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Desmosomal Results: all gene-elusive patients were
mutations: 39 endurance athletes; more intense exerscie,
Gene-elusive 43 p<0.001
Family Hx more often neg in gene-elusive
Exclusion criteria: Gene-elusive patients with most intense
N/A exercise had younger age at presentation,
p=0.025, shorter survival free of VEA, p=0.002
Ruwald AC et al. Study type: North Inclusion: ARVC 1 endpoint: ARVC exercise and VT/VF/SCD Competitive sports associated with
EHJ 2015 (241) Americal ARVC Registry probands. followup 3 y HR: 2.05 for VTA/death and earlier
25896080 registry, 18 centers Results: Patients in competitive sports: presentation of symptoms, c/w
US, Canada Exclusion criteria: Younger at age of Dx, 71% inducible VT/VF, recreational sports or inactive
Age <12 y; ICD >2 y increased risk death/VT.
Size: 108 probands before enrollment;
unknown exercise
level before dx
Sawant AC Heart Study type: Single Inclusion criteria: 1 endpoint: ARVC and outcomes with Recommend restricting unaffected
Rhythm 2016 (242) center retrospective ARVC first degree exercise intensity (MET-HR/y) desmosomal mutation carriers from
26321091 relatives of endurance and high-intensity
Size: 28 probands with PKP2 Results: After adjusting for age, sex, family; athletics, but not from AHA
mutation, interview participation in endurance athletics, (OR: 7.4, recommended minimum levels of
re exercise since p=0.03), higher intensity exercise (OR: 4.2, exercise for heatlhy adults
age 10 y; exercise p=0.004) were associated with dx of ARVCD.
vs AHA
recommendations Family members restricting exercise to 650
to restrict to 390- MET-Hr/yr (AHA upper limits) were sig less
650 MET-HR/y likely to have ARVC dx (OR: 0.07, p=0.002); no
VT/VF
Exclusion criteria:
N/A (AHA/AC Sports Med recommend healthy
adults participate in minimum, 450-750 MET-
min weekly =390650 MET-Hr/y)
Saberniak J et al. Study type: single Inclusion criteria: 1 endpoint: ARVC assess exercise ventricular ARVC athletes showed reduced
Eur J Heart F 2014 center ARVC probands and function with echo, CMR biventricular function compared with
(243) mutation positive Athlete: intensity 6 METS, duration 4 h/wk non-athletes and mutation-positive
25319773 Size: 110 family members Results: Function reduced in athletes vs non- family members
athletes by echo and MRI, all p<0.01.

137

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Genotyping in 100 METs x min/wk correlated with reduced RV Amount and intensity of exercise
patients and LV function p<0.01 was assoc with impaired LV and RV
75% mutation LVEF by MRI reduced in athletes, index and function
positive, PKP 91%, family members Exercise aggravates, accelerates
Syncope 44%, ICD Exercise induced VA in 37% of patients, more myocardial dysfunction in ARVC
47% likely in athletes p<0.001 and in those w
increased duration exercise 2.5 h/wk x 6 y
Exclusion criteria:
N/A
Sen-Chowdry S et Study type: Inclusion criteria: 1 endpoint: ARVC presenting as LV LV dominant ARVC subtype under-
al. JACC 2008 (244) observational ARVC patients w dominant arrhythmogenic cardiomyopathy recognized
19095136 cohort clinical suggestion (LDAC): CMR & clinical Unexplained T wave inversion V5,
of LV involvement: V6 V4, I, aVL
Size: 42 one or more: RBBB Results: Desmosomal mutations present in VT of RBBB morphology,
morphology 45% of probands, 33% of families LV aneurysms
arrhythmia, isolated Arrhythmia of RBBB morphology exceeding LV dilation and/or systolic
(infero) lateral T degree of ventricular dysfunction impairment with arrhythmic
wave inversion, distinguished ARVC from dilated presentation
proven family dx LV cardiomyopathy Extensive LGE of LV myocardium
ARVC or idiopathic inflammatory myocarditis part of
myocardial fibrosis CMR: 88% RV segmental dil and/or wall nat Hx of ARVC
motion abnormality; 27% low RVEF; LV
Clinical eval: involvement 34% dilation or decreased EF.
includes CMR (41
patients): LV late gadolinium enhancement
consensus >2 Inflammatory myocarditis on genetic basis:
readers; echo, 10% prior myocarditis
holter, exercise
test, mutation
screening

Exclusion criteria:
HCM, ischemia,
other structural
heart/lung/systemic
disease

138

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Vermes E et al. Study type: Inclusion criteria: 1 endpoint: Compare ARVC CMR criteria 2010 criteria reduced major +
JACC CV Imaging retrospective Patients referred from 19942010; also, assessed 134 patients minor CMR criteria: from 23.5% to
2011 (245) cohort, single for ARVC evaluation with full diagnostic evaluation for ARVC 6.5%
21414577 center by CMR 20052010
Results: original CMR criteria: 23.5% major; new TFC for CMR improved
Size: 294 Exclusion criteria: using 2010: 6.5% major specificity, but may have reduced
N/A Of 69 patients with major criteria 1994, only sensitivity
23% had major criteria 2010
Of 172 with minor---only 1.1% minor criteria
2010

Also, assessed 10 patients with proven ARVC


on complete evaluation:
4/10 met major criteria, none met minor
Specificity for major/minor criteria: 1994-
78/39%; 2010: 94/96%

te Riele AS et al. Study type: Inclusion criteria: 1 endpoint: ARVC electro-anatomical CMR: basal inferior (94%) and basal
JCE 2013(246) multicenter ARVC mutation correlates CMR, EPS anterior RV (87%) and posterolateral
23889974 retrospective: positive patients Mean followup 6 y LV involvement (80% subepicardial
international undergoing CMR, fat infiltration).
registry ARVC EPS. Results: CMR: abnl RV 96%, biventricular: RV apex involved only in advanced
CMR 74, EPS in 11 52%, LV only: 4%. disease.
Size: 80 patients Epicardial delayed activation
PKP2 83% ACE 41%: VT 67%, approp ICD shock 23%, ACA particularly in perivalvar RV area and
10%. LV posterolat wall.
Exclusion criteria: Arrhythmia free survival lower in patients RVOT involved late in disease.
N/A with more abnormal RV segments
24 patients with advanced structural
abnormalities: 1,5, 10 y arrhythmia free
survival= 57%, 42%, 35%
EPS: scar more extensive in epicardium vs
endocardium, p<0.0001; scar map correlated
with CMR locations:
RV epicardial scar subtricuspid 100%, RV basal
anterior wall 64%

139

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Ablation successful in 18/19 VT: 84% were
from RV; no VT from RV apex
te Riele AS et al. Study type: Inclusion criteria: 1 endpoint: ARVC mutation carriers Presence of mutation alone did not
JACC 2013 (247) prospective registry ARVC mutation undergoing risk stratification: incremental confer arrhythmia risk.
23810894 based carriers without value of ECG, Holter, CMR. ECG & holter abnormalities
sustained VA Mean followup 6 y preceded detectable CMR
Size: 69 abnormalities in ARVC mutation
78%: first degree Results: 78% holter; ECG, CMR in all carriers
relatives 68% asymptomatic at presentation ECG PLUS CMR abnormalities
83% PKP2 Abnormal ECG: 57%, abnormal Holter 26% identify high risk group;
mutations (PVCs >500/24 h, or nonsust VT >100 bpm ? ICD for 1 prevention
Abnormal CMR 30% patients with abnormal Evaluation of cardiac structure
Mean age 2715 y ECG/Holter: 48% had abnormal CMR, vs 4% in and function using CMR is probably
patients with normal ECG/Holter, p<0.0001 not necessary in the absence of
Exclusion criteria: Only 1 pt with normal ECG/holter had baseline electrical abnormalities
ARVC with prior abnormal CMR.
sustained VA Development of sust VA: 16% mean time to
arrhythmia 4.5 y
All patients with sust VA presented with
electrical abnormalities; all had abnormal
CMR.

Patients with both electrical and CMR


abnormalities: higher VA, p <0.0001:
arrhythmia free survival at 1,5,10 y: 89%, 54%,
36%.

Liu T et al. J Study type: Inclusion criteria: 1 endpoint: ARVC: effect of revised TFC on 2010 criteria reduced number of
Cardiovasc magn retrospective cohort patients referred CMR criteria vs 1994 criteria. total patients meeting diagnostic
Reson 2014 (248) 1995-2010 for CMR CMR criteria
24996808 Size: 968 with clinical Results: 2010 criteria reduced no. of total Only 2.6% met diagnostic criteria
suspicion of ARVC patients meeting diagnostic CMR criteria from on CMR
If quantitative RV ~23% to 2.6%: 2.2% met major criteria, 0.4% More objective, quantified criteria
measures not avail, met minor in ARVC dx by CMR
repeat CMR CMR identified alternatic dx in 9.2% of
performed patients, and 4.4% of dx were potential
Mean age 42 y

140

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Males 52% mimics af ARVC-sarcoidosis, other
Exclusion criteria: cardiomyopathies.
N/A
Marcus FI et al. Modifications of Task Force criteria for 1 endpoint: Quantification, specificity of Major criteria
Circ 2010 (249) ARVC ARVC diagnostic criteria. Dysfunction: echo, MRI, angio
20172911 regional dyskinesia, akinesia,
Structural, ECG, arrhythmic and genetic dyssynchrony AND dilation; echo FAC
features as major and minor, with 33%,
quantitative criteria. CMR RVEF 40%; RVEDVI 100
110 ml/m2 (Female/male); localized
SAECG: fQRS fQRSD >114 ms, LASD 38 ms, RV aneurysms or severe segmental
RMS-40 20 V, terminal activation duration dilatiom
QRS 55 ms V1,2, or 3 Tissue bx: residual myocytes
See major criteria at right <60% ECG Repol: age >14 y: Twave
Dx: 2 major, or 1 major plus 2 minor, or 4 inversion V1, V2, and V3;
minor from different groups Depolarization: epsilon V1-3;
Arrhythmia: nonsust/sust VT of
RV fat not part of CMR criteria LBBB, superior axis
Family hx: ARVC confirmed in first
Added mutation status in proband degree relative by TFC, surgery or
autopsy; or pathogenic mutation in
proband
Corrado D et al. Study type: Inclusion criteria: 1 endpoint: ARVC appropr ICD shocks in 1 Overall group had high arrhythmic
Circ 2010 Multicenter consecutive ARVC prevention risk:
20823389 retrospective patients with ICD Mean followup 58 mo
implanted for 1 Univariate analysis: approp shocks:
Size: 106 prevention Results: approp shocks: 24%; inapprop younger, syncope, NSVT, LV
Mean age 36 y shocks 19%; comps 17% dysfunction
Males 67% PES: performed in 60% of patients: 40
Syncope 39% patients (60%) inducible. 65% did not receive Multivar analysis: syncope only
NSVT 53%, family approp therapy; of non-inducible 30% predictor, HR: 3.16, p=0.005
Hx SCD 46% received approp rx. PES PPV 35%, neg PV 70%
Syncope: 43% approp shocks, 4 had recurrent No pt with ICD implanted for
Exclusion criteria: syncope without arrhythmia family Hx only had appropriate
Prior sust VT/VF shocks
Marcus GM et al. Study type: Inclusion criteria: 1 endpoint: Suppression of VEA on AA meds Overall BB not associated with
JACC 2009 Retrospective multi- ARVC patients in in ARVC increase or decrease in VEA;

141

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
19660690 center North Registry treatment Atenolol associated with decreased
American ARVC with ICD and AA Results: risk VEA
Registry drugs BB: used in 61%, (58 patients): no increase or
decrease in VEA; atenolol (20 patients) assoc Sotalol increased risk ICD shock
Size: 95 Exclusion criteria: with decreased risk VEA, HR: 0.25; 95% CI: Amio lower risk VEA
N/A 0.080.80, p=0.018.
Sotalol 38 patients: increased risk ICD shock;
in high dose 320 mg (6 patients) VEA HR: 14.0;
95%CI: 1.6125, p=0.018.
Amio (10 patients) lower risk VEA, HR: 0.25;
95% CI: 0.070.95.
Hershberger RE J Genetic evaluation of Cardiomyopathy Guideline restricts the indication for genetic Details of clinical screening &
Card Fail 2009 (250) testing to that of facilitation of family intervals given:
19254666 screening and management. Ie, Testing is SAECG in ARVC only
used for risk stratification of family members CMR in ARVC
who have little or no clinical evidence of
disease. Recommendations: Childhood: screening intervals
specified relative to ages and
Careful family Hx for 3 generations, for all mutation status
patients.
Especially LMNA mutations
Clinical screening recommended at intervals
for asymptomatic at-risk relatives who are
mutation carriers;

Clinical screening for asymptomatic first


degree relatives when genetic testing has not
been performed/or mutation not identified.

Genetic screening for Fabry disease in all men


w unexplained cardiac disease.

Referral to centers expert in genetic


evaluation and family based management.

142

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Genetic testing for the one most clearly
affected person in a family to facilitate family
screening and management.

ICD may be considered before the LVEF falls


below 35% in patients with CM and significant
arrhythmia or known risk of arrhythmia.

Marcus FI et al. HR Study type: Inclusion criteria: 1 endpoint: Study ARVC clinical eval and Biopsy and CMR least helpful
2009 Multicenter North American diagnostic utility of 7 tests: ECG, SAECG, Diagnostic eval favors: ECG, SAECG,
19560088 retrospective ARVC/D Registry holter, echo, MRI, RV angio, biopsy in 108 echo, RV angio
probands probands referred to core center. Followup Recommend minimum diagnostic
Size: 108 57% male mean 27 mo. eval:
Mean age at dx 38 ECG, SAECG, Holter, echo, RV angio
y Results: 78% of probands classified as
34% competitive affected after evaluation Diagnostic performance of CMR and
athletes Biopsy performed in 59%: should not target biopsy was less than with other tests
Symptoms: ~ all septum but should target RV free wall;
Syncope 21% sarcoidosis found in 3 patients
VA 70% 15% viral infection: Parvovirus 4; enterovirus
Sustained VT 35% not found: ARVC may predispose to viral
Genotype: 100 myocarditis and accelerate disease
patients: 33% progression
positive: PKP2
present in 22% Among 86 patients referred with diagnosis,
23% did not meet TFC, reclassified as
borderline, or not ARVC (2 patients)-mainly
Exclusion criteria: due to CMR interpretation at referring vs core
N/A lab-only 63% confirmed
Choudhary N et al. Study type: Inclusion criteria: 1 endpoint: Presentation, outcomes ARVC No major gender differences in
JCE 2016 Multicenter ARVC probands in by gender outcomes
26840461 Size: 125 North American Mean followup 37 mo Women highest risk age: 31-40 y
ARVC Registry ARVC females: increased PVCs on
Males 56% Results: ACE more likely in affected vs Holter, 2200 vs 1089, p=0.016
109 genotype borderline SAECG: ACE in females-equal in
testing ICD VT/VF or SCD: no difference patients w or w/out abnl SAEC
Exclusion criteria: Fast VT/VF or death in women trend to lower In males, ACE more likely if abnl
N/A risk, HR: 0.41 SAECG
143

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Males: Increase in Abnormal SAECG 81% vs cardiac events not different in
48%, p<0.001, inducible VT/VF 60% vs 40%, genotype positive vs negative
p=0.026

Overall VT/VF shocks: 27% women, 41% men


Genotype positive: 38%, of positive: PKP-2
71%; genotype = gender
2 mutations: 8%

Saguner AM AJC Study type: Inclusion criteria: 1 endpoint: ARVC utility of V-stim to predict study included symptomatic
2013 Prospective single ARVC patients outcomes: positive EP = sustained patients with clinical VT/VF/syncope
23103200 center undergoing EPS monomorphic VT only, triple VEST, =/- isuprel and ventricular dysfunction
NOTE prior to Results: 55% sustained monomorphic VT
Size: 62 study inducible at PES correlated with increased risk Cannot identify how many patients
39% had clinical adverse outcome were asymptomatic with normal
hemodynamically ventricular function
compromised VT Inducibility of sust monomorphic VT (HR:
or VF; 32% sust VT 2.52; 95% CI:1.036.16, p=0.043) and
stable; 50% nonadherence to meds and activity
syncope; restrictions (HR: 2.34; 95% CI: 1.14.99,
NYHA Class II-III p=0.028)
31%; PPV 65%, NPV 71%
LVEF <50% in 24% Anti-tach pacing successfully terminated VT >
RV FAC <33% in 90% of cases
48%
Exclusion criteria:
N/A

144

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 31. Nonrandomized Trials, Observational Studies, and/or Registries of Hypertrophic Cardiomyopathy (Section 7.4)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Maron et al. 2000 Study type: Retrospective, Inclusion criteria: HCM 1 endpoint: ICD shock VT or VF are the principal
(251) multicenter, observational patients at high risk for from VT or VF mechanisms of SCD in HCM
10666426 SCD treated with ICD ICDs are highly effective in high risk
Size: 128 patients Results: At 3.1 y follow up, patients
Exclusion criteria: the ICD delivered
Inadequate data appropriate therapy in 23%
of patients (7%/y). 25% of
patients had an
inappropriate shock.
Therapy for 1 prevention
patients was 5%/y; and for
2 prevention 11%/y.
Christiaans et al. Study type: observational, Inclusion criteria: 1 endpoint: satisfaction The majority of genetic carriers of
2009 (252) single center Predictively tested HCM with genetic counseling HCM gene(s) were satisfied with
19533783 mutation carriers genetic counseling
Size: 143 patients followed by Results: Genetic counseling Receiving information by mail was
questionnaire was valued positively and satisfactory
only 4 carriers would rather
Exclusion criteria: not have known that they
inadequate data were a mutation carrier.
Hamang et al 2012 Study type: Prospective, Inclusion criteria: 1 endpoint: Development Patients with a clinical diagnosis of
(253) multi-center observational Norwegian patients of heart-focused anxiety HCM receiving genetic counseling
21773878 study with a clinical diagnosis continue to experience anxiety.
or genetic risk of HCM Results: 1 y of follow-up Patients with a genetic risk for HCM
Size: 126 patients attending genetic questionnaires after genetic had less anxiety if they experienced
counseling counseling. Patients with a satisfaction with genetic counseling
clinical diagnosis of HCM
Exclusion criteria: compared to genetic risk
inadequate data had higher avoidance
(p<0.002), attention
(p<0.005) and fear
(p<0.007).

145

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Bos JM et al Study type: Single center, Inclusion criteria: 1 endpoint: Genetic Predictors of a positive genetic test
2014 (254) observational data registry Established clinical HCM testing for HCM were reverse curve morphological
24793961 diagnosis subtype, age <45 y, LV wall thickness
Size: 1053 patients Results: 1053 patients with 20 mm, family history of HCM, and
Exclusion criteria: clinical HCM (mean age family history of SCD. Hypertension
Inadequate data 44.419 y) had genetic was not predictive.
testing evaluating 9 HCM- A positive genetic test was predicted
associated myofilament in 6% of patients with only
genes. 34% were positive or hypertension and 80% with all 5
a HCM mutation. . predictor markers.
OMahony et al. Study type: Prognostic Inclusion criteria: HCM 1 endpoint: SCD or Risk modifiers for SCD used in the
2014 (255) model derived from a patients appropriate ICD shock model were age, maximal LV wall
24126876 retrospective, multicenter thickness, left atrial diameter, LV
longitudinal cohort study Exclusion criteria: Results: Median follow-up outflow tract gradient, family Hx of
Clinical risk prediction inadequate data 5.7 y; 5% of patients had SCD, non-sustained VT, and
model for SCD in HCM SCD/ICD shock. 8 pre- unexplained syncope
specified predictors were This is the first validated SCD risk
Size: 3,675 patients associated with SCD/ICD prediction model for patients with
shock at 15% significance HCM and provides accurate
level. Model developed to individualized estimates for the
estimate probability of SCD probability of SCD using clinical
at 5 y. For every 16 ICDs parameters.
implanted in patients with a
4% 5-y SCD risk, potentially
1 pt will be saved.
Elliott et al. 1999 Study type: single center, Inclusion criteria: HCM 1 endpoint: Survival free ICD therapy was better than
(256) observational patients surviving from SCD or appropriate ICD amiodarone at preventing recurrent
10334430 Survival after SCD or resuscitated VF or shock SCD
sustained VT in HCM: syncopal sustained VT Small numbers and purely
treated with amiodarone Results: 8 patients on observational without controls
or ICD Exclusion criteria: amiodarone and 6 received reported.
inadequate data an ICD. Mean follow-up
Size: 16 patients 6.14 y 2 patients on
amiodarone with SCD and 3
patients had appropriate
ICD shock.

146

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Maron et al. 2007 Study type: Inclusion criteria: HCM 1 endpoint: ICD shock ICDs are highly effective in high risk
(257) Retrospective, multicenter, patients at high risk for from VT or VF patients
17652294 registry SCD treated with ICD One death due to VT/VF when ICD
ICD to prevent SCD in HCM Results: 20% had failed to function
Size: 506 patients Exclusion criteria: appropriate treatment of Inappropriate shocks in 27% of
Inadequate data VT/VF: 10.6% per y for 2 patients
prevention and 3.6%/y for A single modifier of high risk for SCD
1 prevention. Time to 1st may be sufficient to justify ICD
appropriate shock was 10 y. placement
Appropriate discharge was
similar in patients with 1, 2,
or 3 risk factors (p=0.77)
Lin G et al. 2009 Study type: Retrospective, Inclusion criteria: 1 endpoint: Inappropriate Inappropriate shocks and device
(258) single center, registry Patients with HCM shocks and device complications are significant in HCM
19282314 Complications and receiving ICD complications patients receiving an ICD
inappropriate ICD shocks Younger patients and those with AF
in HCM patients Exclusion criteria: Results: Mean follow up more likely to have problems
Inadequate data 4.92 y. 36% of patients had
Size: 181 patients complications and 23%
inappropriate shocks (5.3%
per y). Appropriate shocks
4%/y.
Syska et al. 2010 Study type: Retrospective, Inclusion criteria: HCM 1 endpoint: ICD therapy ICD therapy is effective in HCM,
(259) observational, single patients at high risk for and relation to clinical risk although the complication rate is
20132378 center VT/VF treated with ICD profile significant.
Efficacy and complications 1, 2, or more risk modifiers did not
of ICD therapy in HCM Exclusion criteria: Results: Average follow up predict appropriate ICD therapies
Inadequate data 4.6 y. 53.8% of 2
Size: 104 patients prevention patients
received an appropriate
therapy and 16.7% of 1
prevention patients.
Complications:
inappropriate shocks
(33.7%), lead dysfunction
(12.5%), and infections
(4.8%).

147

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
OMahony et al. Study type: Inclusion criteria: HCM 1 endpoint: ICD therapy HCM patients with an ICD are
2012 (260) Retrospective, patients at high risk for and complications exposed to frequent inappropriate
21757459 observational, single VT/VF treated with ICD shocks and implant complications
center, cohort Results: 8% of patients
Efficacy and complications Exclusion criteria: received appropriate shocks
of ICD therapy in HCM Inadequate data (2.3%/y). 16% of patients
received inappropriate
Size: 334 patients shocks (4.6%/y). 18% had
implant complications
(5.1%/y) and 30% had
inappropriate shocks
(8.6%/y).
Melacini et al. 2007 Study type: Retrospective, Inclusion criteria: HCM 1 endpoint: Risk of sudden Medical treatment is not absolutely
(261) single center, patients on AAD death protective against risk of SCD in HCM.
17502652 observational
Pharmacological treatment Exclusion criteria: Results: 10% of patients
to prevent SCD in HCM Inadequate data had SCD over an average of
62 mo: 20% on amiodarone
Size: 173 patients (6/30), 9% on verapamil
(4/46) and BB (7/76), and
0% on sotalol (0/21)
McKenna et al. 1985 Study type: single center, Inclusion criteria: HCM 1 endpoint: SCD, recurrent Amiodarone was better than
(262) observational patients with NSVT on VT conventional medications for
4039188 Improved survival with Holter preventing SCD.
amiodarone in HCM and Results: 24 patients during
VT Exclusion criteria: 1976-1977 had NSVT and Study design was purely observational
inadequate data received conventional AAD:
Size: 86 patients 7 patients had SCD during 3
y follow-up. 21 patients
from 1978-1979 with NSVT
received amiodarone: no
SCD on amiodarone during 3
y follow-up.
Olivotto et al.1999 Study type: Prospective, Inclusion criteria: 1 endpoint: Mortality An abnormal BP response during
(263) single center observational Patients with HCM who exercise in HCM was associated with
10362212 underwent exercise Results: 22% had an CV mortality
testing abnormal BP response (9

148

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Prognostic value of BP with hypotension, 19 with However, the positive predictive
response during exercise in Exclusion criteria: failed BP rise). 4.73.7 y value was only 14%. Negative
HCM Inadequate data follow up, 7% died (3 SCD, 6 predictive value 95%
HF). An abnormal BP
Size: 128 patients response predicted
increased risk for CV
mortality (OR: 4.5; 95% CI:
1.120.1).
Sadoul et al.1997 Study type: Prospective, Inclusion criteria: 1 endpoint: Mortality A normal BP response during
(264) single center observational Patients with HCM who exercise identifies low risk young
9386166 Prognostic value of BP underwent exercise Results: 37% had an patients with HCM.
response during exercise in testing abnormal BP response. An abnormal response had a low
HCM During 4422 mo follow up, (15%) positive predictive value and a
Exclusion criteria: SCD occurred in 12 patients: high (97%) predictive value.
Size: 161 patients Inadequate data 3% in normal BP group and
15% in abnormal BP
response group.
Sorajja et al. 2006 Study type: Single center, Inclusion criteria: HCM 1 endpoint: Survival Patients with HCM and massive LVH
(265) retrospective, longitudinal patients with LVH 30 are at increased risk of SCD, especially
16762758 data base. mm Results: 10-y outcome in the young.
assessed. Survival less than
Clinical implications of Exclusion criteria: general population (77% vs
massive hypertrophy in inadequate data 95%, p<0.001). SCD most
HCM common cause of mortality
in younger patients (overall
Size: 107 patients survival 80%)
Maki et al. 1998 Study type: single center, Inclusion criteria: 1 endpoint: SCD Patients with exercise-related SCD
(266) retrospective, data base Patients with HCM were younger and had smaller
9761089 analysis Results: Mean follow-up 9.4 increases in SBP during exercise.
Hemodynamic predictors Exclusion criteria: y; SCD in 9%. Independent
of SCD in HCM Inadequate data predictors of SCD were a
smaller difference between
Size: 309 patients peak and rest SBP during
exercise (p=0.006), and
higher LV outflow tract
pressure gradient at rest
(p=0.003). Exercise-related

149

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
SCD in 8 patients and
exercise-unrelated SCD in 20
patients (mean age 28 vs 47
y, p<0.05).
Elliott et al. 2006 Study type: Single center, Inclusion criteria: HCM 1 endpoint: SCD LV outflow tract gradient 30 mmHg
(267) retrospective, data base patients with LV was an independent risk modifier for
16754630 LV outflow track outflow tract gradient Results: 31.4% had LV SCD/ICD shock with a 2.4-fold
obstruction and SCD risk in measured outflow tract gradient 30 (p=0.003) increase in the risk of
HCM mmHg, followed median of SCD/ICD shock that is increased if other
Exclusion criteria: 61 mo, 5.9% had SCD, VF, or risk modifiers are present.
Size: 917 patients inadequate data appropriate ICD shock. LV
outflow tract gradient 30 Risk of SCD/ICD shock low (0.37%
mmHg associated with annual risk) if the only risk modifier is
reduced survival free from an increased LV outflow tract gradient
SCD and ICD shock (91.4% vs
95.7%. p=0.004)
Monserrat et al. Study type: Retrospective, Inclusion criteria: HCM 1 endpoint: Sudden NSVT was associates with a
2003 (268) single center, with Holter monitoring cardiac death substantial increased risk of SCD in
12957435 observational young patients with HCM
NSVT and risk for SCD in Exclusion criteria: Results: 19.6% had NSVT. No relationship between duration,
young HCM patients Inadequate data Mean follow up 7040 mo. frequency and rate of NSVT runs and
32 died from SCD, 21 had an adverse events.
Size: 531 patients ICD placed with 4
appropriate shocks. The OR
of SCD in HCM 30 y or
younger was 4.35 (95% CI:
1.5412.28; p=0.006);
compared with 2.16 (95% CI:
0.825.96; p=0.1) in patients
older than 30 y.
Spirito et al. 2000 Study type: Retrospective, Inclusion criteria: HCM 1 endpoint: SCD The cumulative risk of SCD was
(269) single center, patients nearly 0 for a wall thickness of 19 mm
10853000 observational Results: 23 patients (4.8%) or less; and was 40% The sudden death
LVH and risk of SCD in Exclusion criteria: had SCD with a mean follow risk in HCM was increased for a left
HCM Inadequate data up of 6.5 y. The risk of SCD ventricular wall thickness of 30 mm or
increased with wall more.
Size: 480 patients thickness: 0 per 1,000 pt y if

150

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
15 mm or less, to 18.2 per
1,000 pt y if 30 mm or more
(95% CI: 7.337.6).
Elliott et al. 2001 Study type: Retrospective, Inclusion criteria: HCM 1 endpoint: Sudden A wall thickness in HCM of 30+ mm
(270) single center, patients cardiac death was associated with SCD.
11273061 observational Most sudden deaths occur in patients
Severe hypertrophy and Exclusion criteria: Results: 39 patients (6.2%) with a thickness less than 30 mm so the
SCD in HCM Inadequate data had SCD or appropriate ICD presence of other risk factors is
shock; 10 had a wall important
Size: 630 patients thickness of 30 mm or more.
Wall thickness of 30 mm or
more had a higher
probability of SCD or shock:
(RR: 2.07; 95% CI: 1.04.25;
p=0.049)
Elliott et al. 2000 Study type: Retrospective, Inclusion criteria: HCM 1 endpoint: Sudden Risk factors for SCD include NSVT,
(271) single center, patients cardiac death syncope, exercise BP response, family
11127463 observational Hx of SCD, left ventricular wall
Risk factors for SCD in HCM Exclusion criteria: Results: Follow up 3.62.5 y. thickness
Inadequate data The SCD free survival was 2 or more risk factors had a high risk
Size: 368 patients 95% with 0 risk factors, 93% for SCD
for 1, 82% for 2, and 36% for
3. Six y SCD risk was 72%
(95% CI: 56%88%) for 2+
risk factors and 94% (95%
CI: 91%98%) for 1 or 0.
Ackerman et al. Study type: Genetic Inclusion criteria: HCM 1 endpoint: Genetic There is profound heterogeneity in
2002 (272) analysis in unrelated HCM patients consenting to abnormalities HCM
12084606 patients genetic analysis Only1% of unrelated individuals had
Malignant mutations in Results: 4 beta myosin one of the 5 malignant mutations.
HCM Exclusion criteria: heavy chain and one
Inadequate data troponin T gene mutation
Size: 293 patients assessed. 3 of the 293
patients had one of the 5
mutations and all 3 <25 y.
Lopes et al. 2013 Study type: Meta-analysis Inclusion criteria: 1 endpoint: Genetic HCM is a heterogeneous disease.
(273) Studies evaluating mutation

151

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
23674365 Meta-analysis of genetic genetic mutations in The establishment of precise
mutations in HCM HCM Results: Sarcomere gene genotype-phenotype relationships
mutation associated with could not be established
Size: 18 publications, Exclusion criteria: Poor younger age (p<0.0005),
2,459 patients study design family Hx of HCM
(p<0.0005), family Hx of SCD
(p<0.0005) and greater wall
thickness (p=0.03).
Bos et al. 2010 (274) Study type: Multicenter, Inclusion criteria: HCM 1 endpoint: SCD or Patients receiving ICD for 1
21059440 consecutive patients, patients with and appropriate ICD discharge prevention because of a family Hx of
prospective data base, without a family Hx of SCD whether as an isolated risk factor
observational SCD in 1st degree Results: 4.63 y follow up, or combined with other markers,
Family Hx and SCD in HCM relatives who received 25 patients (14%) had an experience rates of appropriate ICD
an ICD. appropriate ICD therapy. discharge comparable to that of other
Size: 177 patients Patients with a family Hx of risk factors.
Exclusion criteria: SCD experience ICDs shocks
Inadequate data at a rate (3.7/100 person-y)
similar to patients with
other risk factors (3.1/100
pt y).
Spirito et al. 2009 Study type: Inclusion criteria: HCM 1 endpoint: Relationship Unexplained syncope was a risk
(275) Observational, prospective patients between syncope and SCD factor for SCD in HCM
19307481 data base entry Patients 40 y with syncope
Syncope and risk of SCD in Exclusion criteria: Results: 205 patients (14%) occurring >5 y before evaluation did
HCM Inadequate data had unexplained or neurally- not show an increased risk of SCD.
mediated syncope. 5.65.2 Neurally mediated syncope was not
Size: 1,511 patients y follow up, 74 patients predictive of SCD
(4.9%) had SCD. Relative risk
of SCD was 1.78 (95% CI:
0.883.51; p=0.08) in
unexplained syncope and
0.91 (95% CI: 0.0 3.83;
p=1.0) in neurally-mediated
syncope.
Maron et al. 2009 Study type: Retrospective, Inclusion criteria 1 endpoint: cause of SCD Athletes confined to United States
(276) registry data Athletes who died CVD was found in 54% of the deaths
19221222 suddenly

152

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Sudden deaths in young Results: Average age 196 HCM was the most common finding
competitive athletes. Exclusion criteria: y. The most common in young athletes experiencing SCD due
inadequate data cardiovascular cause was to a cardiac cause.
Size: 1,866 patients HCM (36%)
Kuck et al. 1988 Study type: observational, Inclusion criteria: 1 endpoint: results of PVS PVS induced VA in 33% of both
(277) single center, consecutive symptomatic and symptomatic and asymptomatic HCM
3280318 Role of PVS in HCM asymptomatic patients Results 11 symptomatic and patients.
with HCM 43 asymptomatic patients.
Size: 54 patients 33% of had inducible rabid
Exclusion criteria: monomorphic or
inadequate data polymorphic VT, VF.
Zhu et al. 1998 (278) Study type: observational, Inclusion criteria: HCM 1 endpoint: results of PVS Sustained polymorphic
9474693 single center, consecutive patients with no Hx of and long term follow-up VT/VFinducible in 1/3 of patients with
Role of PVS in HCM SCD HCM with a low subsequent event rate.
Results: Sustained
Size: 53 patients Exclusion criteria: polymorphic VT or VF
inadequate data induced in 35%. Mean
follow-up 4731 mo: no
events (VT, VF, or ICD shock)
in 34 patients with a
negative PVS, 3 events in 19
patients with positive PVS.
Christiaans et al. Study type: observational, Inclusion criteria: 1 endpoint: diagnosis of At first cardiac evaluation 22.6% of
2010 (279) single center, registry data Asymptomatic carriers HCM, long-term outcome asymptomatic carriers were diagnosed
20019025 The yield of risk of an MYBPC3 gene with HCM
stratification for SCD in mutation Results: Clinical HCM was Risk factors for SCD were frequently
HCM myosin-binding C diagnosed in 53 of 235 present and 11% of carriers could be at
gene mutation carriers; Exclusion criteria: mutation carriers (22.6%). risk for SCD.
focus on predictive inadequate data Women were affected less Predictive genetic testing in HCM
screening than men (15% and 32% families and frequent cardiac
respectively, p=0.003)25 evaluation for the presence of HCM
Size: 245 patients carriers (11%) with one or and risk factors for SCD are justified
more risk factors for SCD until advanced age.
and manifest HCM could be
at risk for SCD.
Olivotto et al. 2008 Study type: Multicenter, Inclusion criteria: 1 endpoint: clinical Screening for sarcomere protein
(280) prospective, cohort Unrelated patients with outcomes related to HCM gene mutations in HCM identifies a

153

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
18533079 Myofilament protein gene HCM with genetic broad subgroup of patients with
mutation screening and testing of the 8 HCM- Results: Mean follow-up 4 increased propensity toward long-term
outcome of patients with susceptibility genes y. 62% of patients had impairment of LV function and adverse
HCM mutations (Myofilament- outcome
Exclusion criteria: positive HCM) and 38% These findings were irrespective of
Size: 203 patients inadequate data were myofilament-negative. the myofilament (thick, intermediate,
Myofilament-positive or thin) involved.
patients at increased risk for
CV death, stroke, Class III or
IV HF (25% vs 7% HR: 4.27;
p=0.008)
Ingles et al. 2013 Study type: Multicenter, Inclusion criteria: 1 endpoint: Identify clinical Family Hx is a key clinical predictor of
(281) retrospective, data base Probands with HCM and variables that can predict a positive genetic diagnosis and has
23598715 analysis genetic testing probands with HCM in direct clinical relevance, particularly in
Clinical predictors of whom a pathogenic the pretest genetic counseling setting.
genetic testing outcomes Exclusion criteria: mutation will be identified Multivariate analysis identified
in HCM inadequate data female gender, increased LV wall
Results: 52% of 265 thickness, family Hx of SCD as being
Size: 265 patients patients had at least one associated with the greatest chance of
mutation. Detection rate identifying a gene mutation.
was higher with positive
family Hx (72 vs 29%,
p<0.0001) and positive
family Hx of SCD (89 vs 59%,
p<0.0001).
Jensen et al 2013 Study type: single center, Inclusion criteria: HCM 1 endpoint: Penetrance of The penetrance of HCM in
(282) observational, data patients and their HCM of child relatives of phenotype-negative child relatives at
23197161 registry relatives with clinical patients with HCM risk of developing HCM was 6% after 12
Penetrance of HCM in screening and y of follow-up.
children and adolescents: a predictive genetic Results: After a mean The finding of phenotype conversion
12-y follow-up study of testing follow-up of 12 y, 2 of the in the mid-20s warrants continued
clinical screening and 36 (6%; 95% CI: 2-18) at-risk screening into adulthood.
predictive genetic testing Exclusion criteria: child relatives who were 42% of the child relatives were non-
inadequate data phenotype negative at carriers, and repeat clinical follow-up
Size: 90 probands and 361 conclusion developed HCM could be safely limited to the remaining
relatives phenotype at 26 and 28 y of children.
age.

154

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Bos JM et al 2013 Study type: Single center, Inclusion criteria: 1 endpoint: Genetic Predictors of a positive genetic test
(274) observational data registry Established clinical HCM testing for HCM were reverse curve morphological
24793961 Characterization of a diagnosis subtype, age <45y, LV wall thickness
phenotype-based genetic Results: 1053 patients with 20mm, family Hx of HCM, and family
test prediction score for Exclusion criteria: clinical HCM (mean age 44.4 Hx of SCD. Hypertension was not
unrelated patients with Inadequate data 19 y) had genetic testing predictive.
HCM evaluating 9 HCM- A positive genetic test was predicted
Size: 1053 patients associated myofilament in 6% of patients with only
genes. 34% were positive or hypertension and 80% with all 5
a HCM mutation. . predictor markers.
Girolami F et al 2010 Study type: Multicenter, Inclusion criteria: 1 endpoint: The presence 4 patients with HCM (0.8% of cohort)
(283) observational data registry Patients with clinical of triple sarcomere gene had triple sarcomere gene mutations
20359594 Clinical features and HCM undergoing mutations The clinical outcome in the 4 patients
outcome of HCM genetic testing included resuscitated SCD in 1; ICD
associated with triple Results: Of 488 unrelated implantation due to risk factors in all 4
sarcomere protein gene Exclusion criteria: index HCM patients, 4 with appropriate shocks in 2; and 3
mutations Inadequate data (0.8%) had triple mutations progressed to end-stage HCM by 4th
and significant events during decade with transplant in 1 and
Size: 488 patients follow up. biventricular pacing in 2.
Hershberger RE J Genetic evaluation of Guideline restricts the Details of clinical screening &
Card Fail 2009 (250) Cardiomyopathy indication for genetic testing intervals given:
19254666 to that of facilitation of SAECG in ARVC only
family screening and CMR in ARVC
management. Ie, Testing is
used for risk stratification of Childhood: screening intervals
family members who have specified relative to ages and mutation
little or no clinical evidence status
of disease.
Recommendations: Especially LMNA mutations

Careful family Hx for 3


generations, for all patients.

Clinical screening
recommended at intervals
for asymptomatic at-risk

155

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
relatives who are mutation
carriers;

Clinical screening for


asymptomatic first degree
relatives when genetic
testing has not been
performed/or mutation not
identified.

Genetic screening for Fabry


disease in all men w
unexplained cardiac disease.

Referral to centers expert in


genetic evaluation and
family based management.

Genetic testing for the one


most clearly affected person
in a family to facilitate
family screening and
management.

ICD may be considered


before the LVEF falls below
35% in patients with CM and
significant arrhythmia or
known risk of arrhythmia.
Klues HG, et al. 1995 Aim: To achieve an Inclusion criteria: Results: In HCM the distribution ofLV
(284) understanding of the true Patients with LV LV wall thickness = 1552 hypertrophy is characteristically
7594106 structural heterogeneity of hypertrophy mm (mean 22.35). asymmetric and particularly
HCM Various patterns of heterogeneous, encompassing most
asymmetric LV hypertrophy possible patterns of wall thickening and
Size: were identified with no single morphologic expression
N=600 patients Hypertrophy involved: considered typical or classic.

156

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
2 left ventricular segments A greater extent of LV hypertrophy
(228 patients [38%]) or was associated with younger age and
3 segments (202 patients more marked mitral valve systolic
[34%]) anterior motion and outflow
1 segment in a substantial obstruction but showed no relation to
number of patients (170 either magnitude of symptoms or
[28%]). gender.

The anterior portion of the


ventricular septum:
most frequently showed
thickening (573 patients
[96%]), and the
predominant site of
hypertrophy in most
patients (492 patients
[83%]).
Adabag AS, et al. Aim: To determine the Inclusion criteria: 1 endpoint: Clincail trigger Patients with extreme hypertrophy
(285) clinical circumstances HCM patients who (wall thickness 30 mm) and those at
17126660 under which HCM is underwent a diagnostic Results: high risk for sudden death were more
identified echocardiography HCM was initially suspected often asymptomatic and identified by
only after the onset of routine or family screenings (p<0.0001
Size: N=711 cardiac symptoms or acute and p=0.004, respectively).
cardiac events in 384
patients.

In 327 patients, HCM was


recognized while patients
were asymptomatic:
225 by routine medical
evaluations,
27 of whom HCM was
recognized during
preparticipation
examinations for
competitive sports or other
activities.

157

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Women, older patients (age
50 years), and those with
outflow obstruction at rest
(gradient 30 mm Hg) were
more likely suspected to
have HCM by virtue of
cardiac symptoms or events
(p<0.0001).
Afonso LC, et al. Aim: To profile the utility At the time of this paper, tissue
2008 and pitfalls of established Doppler-derived strain and 2D strain or
19356516 echocardiographic speckle tracking imaging represent
modalities and discuss the robust and rapidly evolving
evolving role of novel technologies that have advanced our
echocardiographic imaging understanding of regional myocardial
modalities such as tissue mechanics in HCM.
Doppler, Doppler-based Ongoing refinements and additional
strain, 2-dimensional strain research will define the incremental
(speckle tracking imaging), role and clinical utility of these
and 3-dimensional imaging promising techniques, including the
in the assessment of HCM. identification of preclinical disease in
carriers of HCM mutations,
improvement of diagnostic accuracy,
risk stratification, planning therapeutic
strategies, and monitoring treatment.

158

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 32. Nonrandomized Trials, Observational Studies, and/or Registries of Myocarditis (Section 7.5)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Cooper et al.1997 Study type: Inclusion criteria: Giant 1 endpoint: survival Giant cell myocarditis is often fatal
(286) observational, cell myocarditis due to HF and VA
9197214 multicenter data base Results: Rate of death or
Natural Hx of giant-cell Exclusion criteria: cardiac transplantation 89%;
myocarditis inadequate data median survival from onset
of symptoms 5.5 mo.
Size: 63 patients
Kandolin et al. 2013 Study type: Inclusion criteria: giant- 1 endpoint: survival 2/3 of patients with giant-cell
(287) observational, cell myocarditis treated myocarditis are free from severe HF or
23149495 retrospective, single with immunosuppression Results: Transplant-free transplantation on immunosuppression
center survival 69% at 1 y, 58% at 2 59% experience life-threatening VT or
Management of giant- Exclusion criteria: y, 52% at 5y. 59% VF
cell myocarditis with inadequate data, unable experienced sustained VA
immunosuppression to use during follow up and 3
immunosuppression received ICD shocks for VT or
Size: 32 patients VF.

Maleszewski et al. Study type: Inclusion criteria: 1 endpoint: Survival free The risk of disease recurrence and
2015 (288) retrospective, Patients with giant-cell from death, transplant progression is high in giant-cell
25882774 observational, myocarditis surviving >1 y myocarditis treated with
multicenter data base without heart Results: mean age 54.613.9 immunosuppression
Long-term risks in transplantation y, follow up 5.5 y starting 1 y Life-threatening VT or VF occurred in
giant cell myocarditis after diagnosis. 12% died; 23% of patients during long-term follow
Exclusion criteria: 19% transplanted; 23% had up
Size: 26 patients inadequate data, need for 19 episodes of VT or VF
transplantation
WEARIT/BIROAD Study type: Inclusion criteria: 1 endpoint: appropriate The wearable defibrillator was
Feldman et al. 2004 Prospective registries symptomatic HF and EF shock form the wearable successful in defibrillating 75% of events
(289) were combined <0.30 (WEARIT) or defibrillator 24% of patients did not tolerate the
14720148 Use of the wearable patients at high risk for device
defibrillator. SCD after MI or bypass Results: 4 mo follow up. 6 of
surgery (BIROAD) 8 defibrillation attempts
Size: 289 patients successful; 6 inappropriate
159

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: shocks. 6 SCD during study: 5
inadequate data not wearing and 1 incorrectly
wearing device. 68 did not
tolerate vest

Kao et al. 2012 (290) Study type: Inclusion criteria: HF 1 endpoint: sudden death The event rate was too low to allow
23234574 multicenter, patients awaiting assessment of the wearable defibrillator
prospective registry transplantation, dilated Results: 7558 d follow up.
Wearable defibrillator cardiomyopathy, or No episodes of sudden CA.
in HF receiving inotropic
medicines
Size: 82 patients
Exclusion criteria:
inadequate data

Data Supplement 33. Nonrandomized Trials, Observational Studies, and/or Registries of Cardiac Sarcoidosis (Section 7.6)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Naruse et al. Aim: This study Inclusion criteria: 37 1 endpoint: freedom from any VT
2014 (291) sought to describe consecutive patients (11
24837644 both clinical and EP men; age, 5611 y) with a Results: During a 39 mo follow-up, 23
characteristics and diagnosis of sustained VT (62%) patients were free from any VT
outcomes of associated with CS. Clinical episodes with medical therapy. Fourteen
systematic treatment effects of a systematic patients who experienced VT recurrences
approach to VT treatment approach even while on drug therapy underwent
associated with CS. including medical therapy radiofrequency catheter ablation. After a
(both steroid and 33 mo follow-up subsequent to the
Study type: Single antiarrhythmic agents), in radiofrequency catheter ablation, 6 of 14
center observational association with patients experienced VT recurrence. The
radiofrequency catheter number of VTs sustained during EPS was
Size: 37 patients ablation, were evaluated. higher in the patients with VT recurrence
than in those without (3.71.4 vs 1.90.8;
Exclusion criteria: N/A p<0.01).

160

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Takaya Y, et al. Aim: to assess Inclusion criteria: Fifty- 1 endpoint: major adverse cardiac Positive myocardial uptake of 67
2015 (292) outcomes in patients three consecutive patients events, including cardiac death, VF, Ga or 18 F-FDG disappeared after
Am J Cardiol. with AVB as an initial with cardiac sarcoidosis, sustained VT, and hospitalization for HF. the initiation of steroid treatment
2015 Feb 15 manifestation of who had high-degree AVB in all patients, and high-degree
25529542 cardiac sarcoidosis (N=22) or VT and/or HF Results: Over a median follow-up period AVB recovered in some patients,
compared with those (N=31), were enrolled of 34 mo, the outcomes of major adverse indicating that steroid treatment
in patients with VT cardiac events were better in patients was effective but might not be
and/or HF. Exclusion criteria: N/A with high-degree AVB than in those with sufficient for preventing the fatal
VT and/or HF (log-rank test, p=0.046). cardiac events in patients with
Study type: single However, this difference was due mainly high-degree AVB.
center observational to HF hospitalization. The outcomes of
fatal cardiac events, including cardiac
Size: 53 pts death, VF, and sustained VT, were
comparable between the 2 groups (log-
rank test, p=0.877
Kandolin et al. Aim: assess the Inclusion criteria: adult 1 endpoint: serious cardiovascular With current therapy, the
2015 (293) epidemiology, (>18y of age) patients events prognosis of CS appears better
25527698 characteristics, and diagnosed with than generally considered, but
outcome of CS in histologically confirmed CS Results: Altogether, 102 of the 110 patients presenting with HF still
Finland in Finland between 1988 patients received immunosuppressive have poor long-term outcome.
and 2012. A total of 110 therapy, and 56 received an ICD. Left Steroids appeared to stabilize
Study type: patients (71 women) 519 y ventricular function was impaired (LVEF disease but not reverse it. 10-y
Retrospective of age (meanSD) were <50%) in 65 patients (59%) at diagnosis estimate of transplantation-free
found and followed up for and showed no overall change over 12 cardiac survival was as high as 91%
Size: 110 patients outcome events to the end mo of steroid therapy. During follow-up in patients who were diagnosed
of 2013. (median, 6.6 y), 10 patients died of a clinically and received
cardiac cause, 11 patients underwent contemporary immunosuppressive
Exclusion criteria: N/A transplantation, and another 11 patients and device therapy.
suffered an aborted SCD. The KM EF <35% was most important
estimates for 1-, 5-, and 10-y predictor of outcomes
transplantation-free cardiac survival were
97%, 90%, and 83%, respectively. HF at
presentation predicted poor outcome
(log-rank p=0.0001) with a 10 y
transplantation-free cardiac survival of
only 53%.

161

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Yazaki et al. Aim: To determine Inclusion criteria: 95 1 endpoint: predictors of mortality Authors concluded that the
2001 (294) the significant Japanese patients with CS. severity of HF was one of the most
11703997 predictors of Twenty of the 95 patients Results: During the mean follow-up of 68 significant independent predictors
mortality and to had never received mo, 29 patients (73%) died of CHF and 11 of mortality for CS. Starting
assess the efficacy of corticosteroid therapy (27%) experienced sudden death. KM corticosteroids before the
corticosteroids because the sarcoidosis had survival curves showed 5-y survival rates occurrence of systolic dysfunction
not been diagnosed before of 75% in the steroid-treated patients resulted in an excellent clinical
Study type: their deaths; sarcoidosis and of 89% in patients with a LVEF 50%, outcome
retrospective was proved at autospy. The whereas there was only 10% 5 y survival
multicenter in Japan other 75 patients treated rate in autopsy subjects. Multivariate
with corticosteroids were analysis identified NYHA functional class
Size: 95 patients classified into 2 cohorts HR: 7.72 per class I increase, p=0.0008),
according to initial LVEF left ventricular end-diastolic diameter
obtained by contrast left (HR: 2.60/10 mm increase, p=0.02), and
ventriculography or sustained VT (HR: 7.20, p=0.03) as
echocardiography: LVEF independent predictors of mortality.
50% (N=39) or LVEF <50%
(36).

Exclusion criteria: N/A


Aizer A, et al. Aim: To evaluate the Inclusion criteria: 1 endpoint: appropriate ICD therapies or Most patients had syncope, NSVT
2005 (295) utility of Consecutive patients with sudden death or presysncope and mean EF in the
Am J Cardiol. programmed cardiac sarcoidosis inducible was 33.217.0
2005 ventricular underwent programmed Results: 5 of 6 patients (83%) with
16018857 stimulation to predict ventricular stimulation. spontaneous sustained ventricular
future arrhythmic Patients with spontaneous arrhythmias and 4 of 6 patients (67%)
events in patients or inducible sustained without spontaneous but with inducible
with cardiac ventricular arrhythmias sustained ventricular arrhythmias
sarcoidosis (N=12) underwent ICD received appropriate ICD therapy. 2 of 20
insertion patients (10%) with neither spontaneous
Study type: Single nor inducible sustained ventricular
center Exclusion criteria: NA arrhythmias experienced sustained
ventricular arrhythmias or sudden death.
Size: 32 pts Programmed ventricular stimulation
predicted subsequent arrhythmic events
in the entire population (relative HR:
4.47; 95% CI: 1.3015.39) and in patients

162

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
who presented without spontaneous
sustained ventricular arrhythmias
(relative HR: 6.97; 95% CI: 1.2738.27).
Mehta D., et al. Aim: to assess the Inclusion criteria: Patients 1 endpoint: survival and arrhythmic Authors mention that based on
2011 (296) value of programmed with biopsy-proven events. present clinical indications, a
Circ Arrhythm electric stimulation of systemic sarcoidosis but significant proportion of patients
Electrophysiol. the ventricle (PES) for without cardiac symptoms Results: Eight (11%) were inducible for with CS and LVEF of <35% would
2011 risk stratification in who had evidence of sustained VA and received an ICD. None qualify for ICD implantation. There
21193539 patients with cardiac sarcoidosis on PET of the noninducible patients received a are no data to guide management
sarcoidosis or CMR were included defibrillator. LVEF was lower in patients of patients with minimal or mild LV
Study type: Single with inducible VA (36.44.2% vs dysfunction who lack evidence of
center 1998-2008 Exclusion criteria: prior 55.81.5%, p<0.05). Over a median VA or conduction system disease.
history of ventricular follow-up of 5 y, 6 of 8 patients in the
Size: 76 pts arrhythmias or ICD group with inducible VA had VA or died,
compared with 1 death in the negative
group
Coleman et al. Aim: This study Inclusion criteria: Studies 1 endpoint: all-cause mortality and a This analysis shows that the
2016 (297) sought to perform a were considered eligible for composite outcome of arrhythmogenic presence of LGE in sarcoid patients
27450877 systematic review inclusion if events plus all-cause mortality. with normal or near-normal LVEF is
and meta-analysis to CMR was used to assess for prognostically significant and
understand the myocardial scarring from Results: The average EF was 57.89.1%. greatly increases the likelihood of
prognostic value of biopsy-proven or clinically Patients with LGE had higher odds for all- adverse events.
myocardial scarring suspected sarcoidosis; in cause mortality (OR: 3.06; p<0.03) and
as evidenced by late cohorts of >5 patients; with higher odds of the composite outcome
gadolinium >1 y of prognostic (OR: 10.74; p<0.00001) than those
enhancement (298) follow-up data, including without LGE. Patients with LGE had an
on CMR imaging in event data for ventricular increased annualized event rate of the
patients with known arrhythmia, SCD, aborted composite outcome (11.9% vs. 1.1%;
or suspected CS. cardiac death and/or p<0.0001).
appropriate ICD discharge,
Study type: Meta hospital admission for
analysis congestive HF, cardiac
mortality, and allcause
Size: Ten studies mortality.
were included,
involving a total of Exclusion criteria: Studies
760 patients. with populations known to

163

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
have CAD or
cardiomyopathies of
nonsarcoid etiology.
Murtagh et al. Aim: The aim of this Inclusion criteria: 205 1 endpoint: death or any VT The burden of LGE and the
2016 (299) study was to patients with LVEF >50% severity of RV dysfunction further
26763280 establish whether and extracardiac sarcoidosis refine the risk of death/VT in
CMR with LGE who underwent Results: Forty-one of 205 patients (20%) patients with CS
imaging can be used cardiovascular magnetic had LGE; 12 of 205 (6%) died or had VT
to risk stratify resonance for LGE during follow-up; of these, 10 (83%) were
patients with known evaluation in the LGE+ group. In the LGE+ group (1)
extracardiac the rate of death/VT/y was >20 higher
sarcoidosis and Exclusion criteria: N/A than LGE- (4.9 vs. 0.2%, p<0.01); (2)
preserved LVEF death/VT were associated with a greater
(>50%). burden of LGE (1411 vs. 55%, p<0.01)
and right ventricular dysfunction (right
Study type: Single ventricular EF 4512 vs. 5328%, p=0.04).
center retrospective LGE burden was the best predictor of
death/VT (area under the receiver-
Size: 205 patients operating characteristics curve, 0.80); for
every 1% increase of LGE burden, the
hazard of death/VT increased by 8%.
Crawford et al. Aim: to assess Inclusion criteria: Fifty-one 1 endpoint: death or VT/VF A cut-off value of 9 involved
2014 (300) whether delayed patients with CS and LVEF segments separated patients with
25266311 enhancement (DE) on >35% underwent DE-MRI. and without future VTs, suggesting
MRI is associated DE was assessed by visual Results: Twenty-two of 51 patients (63%) that a threshold effect may be
with VT/VF or death scoring and quantified with had DE. Forty patients had no prior Hx of present. Right ventricular
in patients with CS the full-width at half- VT (1 prevention cohort). Among those, involvement seems to be
and LVEF>35%. maximum method. The 3 patients developed VT and 2 patients particularly important for
patients were followed for died. DE was associated with risk of arrhythmogenesis; it was
Study type: 48.020.2 mo. VT/VF or death (p=0.0032 for any DE and predictive of adverse events in 1
Retrospective p<0.0001 for right ventricular DE). The prevention patients and for the
analysis from Exclusion criteria: N/A positive predictive values of the presence group as a whole. Patients without
multicenter registry of any DE, multifocal DE, and right DE on MRI have a low risk of VT.
ventricular DE for death or VT/VF at
Size: 51 patients mean follow-up of 48 mo were 22%, 48%,
and 100%, respectively.

164

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Greulich et al. Aim: study aimed to Inclusion criteria: 155 1 endpoint: 1 endpoints were death, Could not tell on additional LGE
2013 (186) demonstrate that the consecutive patients with aborted SCD, and appropriate ICD parameters due to low numbers.
23498675 presence of late systemic sarcoidosis who discharge.
gadolinium underwent CMR for workup
enhancement (298) is of suspected cardiac Results: LGE was present in 39 patients
a predictor of death sarcoid involvement. The (25.5%). The presence of LGE yields a Cox
and other adverse median follow-up time was HR: 31.6 for death, aborted SCD, or
events in patients 2.6 y. appropriate ICD discharge, and of 33.9
with suspected CS for any event. This is superior to
Exclusion criteria: N/A functional or clinical parameters such as
Study type: LVEF, LV end-diastolic volume, or
Multicenter presentation as HF, yielding HRs between
prospective 0.99 (per % increase LVEF) and 1.004
(presentation as HF), and between 0.94
Size: 155 patients and 1.2 for potentially lethal or other
adverse events, respectively.
Blankstein et al. Aim: to relate Inclusion criteria: 1 endpoint: Death or VT Conclusion was that presence of
2014 (301) imaging findings on consecutive patients with focal PD and FDG uptake on cardiac
24140661 positron emission no Hx of CAD, who were Results: Among the 118 patients (age PET identifies patients at higher
tomography (PET) to referred for PET, using 5211 y; 57% males; mean EF: 4716%), risk of death or VT.
adverse cardiac (18)F-fluorodeoxyglucose to 47 (40%) had normal and 71 (60%) had
events in patients assess for inflammation and abnormal cardiac PET findings. Over a
referred for rubidium-82 to evaluate for median follow-up of 1.5 y, there were 31
evaluation of known perfusion defects (PD), (26%) adverse events (27 VT and 8
or suspected CS. following a high-fat/low- deaths). Cardiac PET findings were
carbohydrate diet to predictive of AE, and the presence of
Study type: Single suppress normal myocardial both a PD and abnormal FDG (29% of
center observational glucose uptake patients) was associated with HR:3.9;
p<0.01 and remained significant after
Size: 118 patients Exclusion criteria: N/A adjusting for LVEF and clinical criteria.
Extra-cardiac FDG uptake (26% of
patients) was not associated with AE.
Kron et al. 2013 Aim: to evaluate the Inclusion criteria: 1 endpoint: appropriate ICD therapy Patients receiving appropriate
(302) efficacy and safety of consecutive patients with therapies were more likely to be
23002195 ICDs in patients with CS and an ICD at 13 male, have a Hx of syncope, have a
CS academic centers. Results: Over a mean follow-up of lower LVEF, a 2 prevention ICD
4.24.0 y, 85 of 234 (36.2%) patients indication

165

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: 147 patients (62.6%) had received an appropriate ICD therapy Most patients receiving
multicentre their devices implanted for (shocks and/or anti-tachycardia pacing) appropriate therapies had an LVEF
retrospective data 1 prevention while 88 and 67 of 226 (29.7%) received an >35%, suggesting that CS patients
review patients (37.5%) were appropriate shock. with mild or moderately reduced
implanted for 2 LVEF may be at risk for VA
Size: 235 patients prevention, including 7 for
from 13 institutions VF (3.0%), 63 for VT
(26.8%), and 18 for syncope
presumed to be due to an
arrhythmia (7.7%).

Exclusion criteria: N/A


Mohsen et al. Aim: to identify the Inclusion criteria: Patients 1 endpoint: appropriate ICD therapy CS is strongly associated with
2014 (303) predictors of life- with biopsy-proven malignant VA. No specific
24433308 threatening VA in systemic sarcoidosis but predictors of such
patients with CS and without cardiac symptoms Results: The mean LVEF was 4118%. tachyarrhythmias emerged, other
to evaluate the role who had evidence of CS on Thirty patients received an ICD. Twelve than young age and low LVEF.
of the ICD in this positron emission patients (36.3%) had sustained VA. Over 2/3 received ICD for 2
patient population. tomography (PET) or CMR Eleven patients received appropriate prevention
were included therapies and 9 patients received
Study type: inappropriate shocks, representing 36.7%
multicentre Exclusion criteria: N/A and 30.0% of the ICD population,
retrospective data respectively. Patients who received
review appropriate ICD therapies were younger
with mean age 47.47.8, and had a lower
Size: 32 patients. mean LVEF 33.012.0 compared to those
84% received the ICD who did not receive ICD therapies
for symptoms. (p=0.0301 and 0.0341, respectively).
Schuller et al. Aim: identify the Inclusion criteria: Patients 1 endpoint: Any ICD therapy Appropriate ICD therapies were
2012 (304) incidence and with CS and an ICD higher than in historical control
22812589 characteristics of ICD implanted for 1 or 2 Results: Of the 112 CS subjects identified,
therapies in patients prevention of sudden 36 (32.1%) received appropriate
with CS death. Additionally, authors therapies VT over a mean follow-up
included a comparison with period of 29.2 mo. VT storm (>3 episodes
Study type: historical controls of ICD in 24 h) occurred in 16 (14.2%) CS
multicentre therapy rates reported in subjects. Inappropriate therapies
observational clinical trials evaluating the occurred in 13 CS subjects (11.6%).

166

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
ICD for 1 and 2 prevention Covariates associated with appropriate
Size: 32 patients. of sudden death. ICD therapies included LVEF <55% (OR
84% received the ICD 6.52; 95% CI: 2.4317.5), right ventricular
for symptoms. Exclusion criteria: N/A dysfunction (OR: 6.73; 95% CI: 2.69
16.8), and symptomatic HF (OR: 4.33;
95% CI: 1.8610.1).
Yodogawa et al. Aim: to evaluate the Inclusion criteria: Patients 1 endpoint: PVCs and NSVT burden Steroid therapy may be effective
2011 (305) efficacy of presenting premature before and after steroid therapy. for VA in the early stage, but less
21496164 corticosteroid ventricular contractions effective in the late stage
therapy VA in CS (PVCs 300/d) were Results: The group with less advanced LV
investigated. All were dysfunction patients (EF 35%, N=17)
Study type: Single treated with steroids. showed significant reduction in the
center observational number of PVCs (from 18202969 to
Exclusion criteria: N/A 7421425, p=0.048) and in the
Size: 31 patients prevalence of NSVT (from 41 to 6%,
p=0.039). Late potentials on SAECG were
abolished in 3 patients. The less
advanced LV dysfunction group showed a
significantly higher prevalence of gallium-
67 uptake compared with the advanced
LV dysfunction group (EF <35 %, N=14). In
the advanced LV dysfunction patients,
there were no significant differences in
these parameters.
Segawa et Aim: to evaluate Inclusion criteria: Patients 1 endpoint: Sustained VA. These results indicate that VTs
al.2016 (306) time course and presenting with CS treated and electric storm frequently occur
27301264 factors correlating with steroids. Results: During a mean follow-up of 5.5 in the first 12mo after initiation of
with VT after y, 20 out of 68 patients (29%) corticosteroid therapy, presumably
introduction of Exclusion criteria: N/A experienced VTs after initiation of because of inflammatory
corticosteroid corticosteroid therapy, especially in the conditions, and that the positive
therapy in patients first 12 mo in 14 patients (70%). A gallium scintigraphy is a significant
with CS remain to be multivariable analysis revealed that and independent predictor of VTs
elucidated. positive gallium scintigraphy had a
significant correlation with VTs (HR:
Study type: Single 11.33; 95% CI: 3.2239.92; p<0.001), in
center observational addition to reduced LVEF (HR: 0.94; 95%
CI: 0.900.97; p=0.001). Furthermore,

167

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 68 patients electrical storm was noted in 10 patients
(14.7%), 8 within the first 12mo of
treatment, whereas the recurrence of
electric storm was relatively less.

Data Supplement 34. Nonrandomized Trials, Observational Studies, and/or Registries of Other Infiltrative Cardiomyopathies (Section 7.6.1)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Varr et al. 2014 (307) Aim: To test whether Inclusion criteria: The 1 endpoint: VA Of the 6 patients who received ICD
24121001 there is a specific Stanford Amyloid therapies, 4 died within 18 mo and 3
population of patients Centers database to Results: NSVT was common received the ICD initially for 1
with cardiac identify all patients with and occurred in 23 of 31 (74%) prevention.
amyloidosis at risk of AL or ATTR who had patients. Sustained VT or VF The authors proposed criteria for ICD
SCD owing to VA (vs ambulatory cardiac occurred in 6 of 31 (19%) implant
EMD) who would monitoring. This included patients over the study That included syncope, VT or NSVT.
benefit from ICD patients who had period. Of the 6 patients with
undergone interrogation VT/VF, 1 patient had
Study type: of an ICD or pacemaker spontaneous resolution of VT
Retrospective registry and those who had before the delivery of ICD
Database analysis ambulatory monitoring in therapy. The remaining 5
the outpatient setting patients had ICD therapies
Size: 31 with either a Holter used, either antitachycardia
monitor or Ziopatch pacing (ATP) or defibrillation.
(iRhythm technologies, All patients had had
San Francisco, CA). documented NSVT before ICD
therapy for VT/VF.
Exclusion criteria:
patients who did not
have any form of
telemetry monitoring
available
Kristen et al. 2008 Aim: to test whether Inclusion criteria: 1 endpoint: mortality Authors concluded that patients with
(308) prophylactic placement patients with cardiac amyloidosis predominantly die as
168

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
18242546 of an ICD reduces SCD histologically proven Results: During a mean a result of electromechanical
in patients with cardiac cardiac amyloidosis and follow-up of 811151 d, 2 dissociation and other diagnoses not
amyloidosis risk of sudden death as patients with sustained VT amenable to ICD therapy. Selected
demonstrated by a Hx of were successfully treated by patients with cardiac amyloidosis may
Study type: Single syncope and/or the ICD. Two patients benefit from ICD placement.
center observational ventricular extra beats underwent heart
(Lown grade IVa or transplantation, and 7
Size: 19 higher) patients died due to
electromechanical
Exclusion criteria: N/A dissociation (N=6) or
glioblastoma (N=1).
Lubitz et al. 2008 Study type: Review Inclusion criteria: 1 endpoint: NA Data on sudden death prevention in
(309) Article on SCD in Review article on diseases other than sarcoidosis is very
18634918 infiltrative infiltrative Results: It is difficult to draw scant
cardiomyopathies: cardiomyopathis and substantive conclusions
sarcoidosis, sudden death. Studies regarding the appropriate risk
scleroderma, related to sudden death stratification and therapy of
amyloidosis, and sudden death patients with the infiltrative
hemachromatosis. prevention were cardiomyopathies. Few
discussed. studies are prospective, many
Size: NA use different diagnostic
Exclusion criteria: N/A criteria, and therapies are
rarely randomized.
Furthermore, sample sizes are
small, studies are typically
single center, and the
heterogeneity of disease
manifestations may preclude
the generalization of results.
Patients in high-risk groups,
especially those with
significantly reduced left
ventricular function may be
best treated with prophylactic
ICD.

169

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 35. Nonrandomized Trials, Observational Studies, and/or Registries of Use of ICD and WCD in Patients with HFrEF - (Section
7.8.1)

Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Gandjbakhch E, et al. Study type: single Inclusion criteria: 1 endpoint: all-cause Conclusion: Patients with ICD were
2016 (157) center retrospective consecutive patients mortality less likely to die on the waiting list but
27344378 observational study listed for heart this did not appear in the multivariable
transplantation at 1 Results: Patients with ICD were model to be independently associated
Size: 380 patients center. ICD patients less likely to die on the waiting with mortality.
(122 with ICD) characterized as list (8.3% ICD patients and 19.0%
having ICD before or non-ICD, p=0.001). However, in
within 3 mo after multivariable model, ICD did not
being listed for heart remain an independent
transplant predictor.

Exclusion criteria: N/A ICD-related complications 21%


of patients of which 11.9% was
post-op worsening of HF.

Frohlich GM, et al. Study type: Inclusion criteria: 1 endpoint: all-cause Conclusion: ICD appears to be
Heart 2013 (156) retrospective consecutive patients mortality associated with a reduction in all-cause
23813845 observational study listed for heart mortality compared to those without
transplantation in two Results: estimated 1 y survival an ICD on the waiting list
. Size: 1089 tertiary centers 88% ICD vs. 77% without ICD
consecutive patients (p=0.0001).
listed for heart Exclusion criteria: N/A
transplantation of Model adjustment suggested
which 550 (51%) with ICD independently associated
ICD (216 1 and 334 2 with survival most pronounced
prevention indcations) for those with 1 prevention
indication (HR: 0.4; 95% CI:
0.190.85; p=0.016)

170

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Sandner SE, et al. 2001 Study type: Inclusion criteria: 1 endpoint and results: Total Limitations: retrospective, older
(310) Retrospective Consecutive patients mortality while waiting for study with MADIT I and MUSTT type
11568051 observational study listed for heart transplant was 13.2% with ICD indications for ICD and ICD patients
transplant 1/1992 and and 25.8% without ICD (p=0.03). were highly selected introducing
Size: 854 patients on 3/2000 confounding and baseline clinical
the waiting list for Rate of 12 mo sudden death was variables were not comparable. Low
heart transplant (102 Exclusion criteria: 20% in the non-ICD group and use of BB.
patients with ICD, N/A 0% in the ICD group. Conclusions: supports the use of ICD
11.9%). All patients for improving survival to transplant
had ICD implanted Patient demographics: Cox proportional hazard model
before listing for Indication for ICD was showed absence of ICD
transplant SCA (63%), associated with increased
mortality and sudden death.
60% non-ischemic
etiology

Only 24% overall were


on BB
Kao AC, et al. 2012 Study type: Inclusion: WCD Device worn for 7558 d. 4 Conclusions: WCD monitored HF
(290) Observational prescribed for either patients were on inotropes. patients until further assessment of
23234574 multicenter cohort listed for cardiac There were no sudden cardiac risk. The leading reasons for end of
study transplantation, arrests or deaths during the WCD use were improvement in LVEF or
diagnosed with dilated study. ICD implantation if there was no
Size: 82 cardiomyopathy, or significant improvement in LVEF.
receiving inotropic 41.5% of patients were much
medications. improved after WCD use, while
34.1% went on to receive an
Mean age 56.813.2, ICD.
and 72% were male.
Most patients (98.8%)
were diagnosed with
DCM with a low EF
(<40%) and 12 were
listed for cardiac
transplantation.

171

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Opreanu M et al. 2015 Study type: registry of Inclusion: patients The patients wore the WCD for Conclusions: A significant
(311) patients awaiting awaiting heart an average of 127392 d proportion of patients on the heart
26094085 heart transplant with transplant with WCD (median 39d) with average daily transplant waiting list will have VA.
WCD use of 177 h (median 20h). WCD use in this registry associated with
Seven patients (6%) received a high compliance and efficacy and a
Size: 121 patients appropriate WCD shocks. Fifty- low complication rate, suggesting that
one patients (42%) ended use the WCD is a reasonable bridge therapy
Patient Demographics: after ICD implantation and 13 for preventing SCD in patients awaiting
consisting of 83 (69%) patients (11%) after HT. There HT.
men and 38 (31%) were 11 deaths (9%).
women. The mean age
was 4418 y. Mean EF
was 25 15%. Non-
ischemic
cardiomyopathy (CMP)
was the underlying
diagnosis in 67 (55%)
patients, whereas 21
(17%) patients had
ischemic CMP and 33
(27%) had a mixed or
uncharacterized CMP.
NYHA Class III HF was
present in 32% and
34% were in Class IV.

172

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 36. Nonrandomized Trials, Observational Studies, and/or Registries Related to LVAD (Section 7.8.3)
Study Acronym; Author; Aim of Study; Patient Population Study Intervention Endpoint Results
Year Published Study Type; (# patients) / (Absolute Event Rates,
Study Size (N) Study Comparator P values; OR or RR; &
(# patients) 95% CI)
Vakil, et al. JACCCEP Study type: Inclusion criteria: Adults 1 endpoint: all-cause waitlist Conclusion: ICD use was
2016 (312) retrospective national (age 18 y) listed for first- mortality. associated with improved survival
27395347 registry time HT in the United on the HT waitlist in patients with
States between January 1, Results: 9% died on the wait or without LVADs
Size: 32,599 patients 1999, and September 30, list in ICD group vs. 15% in
2014, were retrospectively no-ICD group (p<0.0001),
identified from the United
Network for Organ Sharing An ICD at listing was
registry. associated reduction in
mortality (HR: 0.87; 95% CI:
Median follow-up of 154 d, 0.800.94).
3,638
In the subgroup of patients
with LVAD (N=9,478), having
an ICD was associated with
relative reduction in mortality
(HR: 0.81; 95% CI 0.700.94).

Data Supplement 37. Nonrandomized Trials, Observational Studies, and/or Registries Related to ICD Use After Heart Transplantation
(Section 7.8.4)
Study Acronym; Aim of Study; Patient Population Study Intervention (# patients) / Endpoint Results
Author; Study Type; Study Comparator (# patients) (Absolute Event Rates,
Year Published Study Size (N) P values; OR or RR; & 95% CI)
Tsai et al. 2009 Study type: Inclusion criteria: 1 endpoint: Descriptive: Use of ICDs after heart
(313) Retrospective cohort of Patients with heart Indications for ICDs and shocks transplantation may be
19808340 Heart Tx. Patients with transplants and ICDs (appropriate/inappropriate) appropriate in selected high-risk
ICDs across 5 centers. patients.
1995-2005 Exclusion criteria: N/A Results: Very small number, no control
indications for ICD group, Pre-SCD-HeFT.
1) severe allograft vasculopathy
(N=12),
173

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 36 (2612 patients 2) unexplained syncope (N=9),
with heart transplants, 36, 3) Hx of CA (N=8),
with ICDs) 4) severe LV dysfunction (N=7).

Shocks: 22 shocks in 10 patients


(28%),
Appropriate: 8 patients/12 shocks
(100% - allograft vasculopathy)
Inappropriate: 3 patients of whom
8 (80%) received 12 appropriate
shocks for either rapid VT or VF.
The shocks were effective in
terminating the
VA in all cases. Three (8%) patients
received 10 inappropriate shocks.
McDowell et al. Study type: Survey of Inclusion criteria: 1 endpoint: Indication, Most common reason was
2009 (314) transplant program Survey responses about allograft vasculopathy with LV
19632584 directors. Asked about all heart transplant patients. Results: dysfunction
transplant patients with an With ICDs Indication for implant*
ICD 1 VT/VF arrest 6 (13.3)
Exclusion criteria: N/A Unexplained syncope 3 (6.7)
Size: 44 patients with CAV with LV dysfunction 20
heart transplants with ICD (44.4)
CAV without LV dysfunction 3
(6.7)
Non-specific graft dysfunction
5 (11.1)
High-grade arrhythmia
determined by
Non-invasive monitor 3 (6.7)
Patients with appropriate
therapies 6 (13.6); Total 19
Patients with inappropriate
therapies 3 (6.8) Total 15
Neylon et al. 2016 Study type: Single center Inclusion criteria: 1 endpoint: Descriptive ICDs in transplant patients
(315) review of transplant inconclusive.
26856670 patients with ICDs Results:
174

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Review of all transplant Allograft vasculopathy in 8/10
Size: 10 patients patients with ICDs 1/10 shocked,
between 1983 and 2012. 1/10 ATP

Exclusion criteria: N/A

Data Supplement 38. Nonrandomized Trials, Observational Studies, and/or Registries Evaluating the Risk of Sudden Death or Ventricular
Arrhythmias in Patients with Neuromuscular Disorders (Section 7.8)
Study Acronym; Study Type/Design; Study Size (N); Patient 1 Endpoint and Results Summary/Conclusion
Author; Study Size Population (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Tanawuttiwat Study type: Observational Inclusion criteria: 136 1 endpoint: Conduction Prevalence of critically prognostic
T, et al. 2017 retrospective cohort referred patients with DM1 and abnormalities were defined as PR conduction abnormalities >20% and LV
(316) for risk stratification at a 28 patients with DM2 of at least 240 msec and QRS of dysfunction > 10% (defined LVEF <55%)
27829084 single referral center with genetically at least 120 msec Incident QRS prolongation > 10 ms is
confirmed diagnosis associated with decreased LV function
Size: 155 patients and baseline ECG Results: In DM1, incidences of PR the subsequent year.
between January 1997 240 ms and QRS 120 ms during Supports serial ECG examinations and
and August 2014. a mean 5.54 y were 19.2% and symptom / QRS prolongation
11.7%, respectively. prompted evaluation of LV function.
Exclusion criteria: Limitations include retrospective
Exclusion of ECGs with In contrast, DM2 patients there design with potential for selection bias,
paced or non-sinus were no incident PR differential clinical follow-up among
rhythm abnormalities, despite similar subgroups.
incidence of QRS abnormalities.

An incident 10 ms increase in
QRS duration was associated with
3.5% decrease in EF in the
subsequent year (3.45; 95% CI:
4.872.03; p<0.001).
Merino et al. Aim: To assess the Inclusion: Consecutive 1 endpoint: N/A Summary A high clinical suspicion
1998 (317) mechanism of sustained VT in patients with myotonic for bundle-branch reentry tachycardia
9714111 myotonic dystrophy dystrophy and Results: Clinical tachycardia was is reasonable in patients with wide
inducible in all patients and were
175

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: Case series sustained VT referred bundle branch reentry. VT was complex tachycardia and myotonic
for EPS no longer inducible after bundle dystrophy
Size: 6 patients branch ablation except for a Limitations small case series. Does
Exclusion: N/A nonclinically documented and not prove a link between bundle
NSVT in a patient with SHD branch reentry and sudden death in
this population

Diegoli et al. Aim: To describe the outcome Inclusion: 1/1995 1 endpoint: N/A DYS-related DCM is characterized by
2011 (318) of patients with dilated 12/2009, screened DYS severe impairment of LV function,
21851881 cardiomyopathy and DYS in 436 unrelated male Results: Of the 34 affected marked LV dilation, and low
defects probands diagnosed patients, 8 patients underwent arrhythmogenic risk; the only factor
with DCM who were heart transplant and 8 patients that impacts survival seems to be end-
Study type: Cohort study male sex received an ICD (indications stage HF.
depressed LVEF). There were no Limitations: relatively small number
Size: 34 patients with DYS Exclusion: females, appropriate interventions during of patients and short follow-up, referral
defects families with male to a median follow-up 14 mo (IQR center.
male transmission 525 mo).
Anselme et al. Aim: To evaluate a strategy of Inclusion criteria: 1 endpoint: N/A Life-threatening VAs are common in
2013 (208) prophylactic ICD implantation LMNA mutation patients with lamin A/C mutations and
23811080 in lamin A/C mutation carriers carriers seen between Results: significant cardiac conduction
with significant cardiac 3/1999 and 4/2009 In those with ICD, 11/21 (52%) disorders, even if LVEF is preserved.
conduction disorders 47 patients (mean had appropriate ICD therapy ICD is an effective treatment and
age 3811 y; 26 men) during a median follow-up of 62 should be considered in this patient
Study type: Cohort study, with LMNA mutation. mo population.
single center 21 (45%) had LVEF was 45% in 9/11 patients
significant conduction with appropriate therapy
Size: 47 patients disorders (defined as The presence of significant
bradycardia requiring conduction disorders is
pacemaker or a PR associated with malignant VA
interval of >240 ms and (HR: 5.20; 95% CI: 1.1423.53;
either complete LBBB p=0.03)
or NSVT) and received
a prophylactic ICD

Exclusion criteria: N/A

176

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
van Rijsingen Aim: To identify risk factors Inclusion criteria: 1 endpoint: Occurrence of Patients with lamin A/C mutations
et al. 2012 (209) that predict malignant VAs in Pathogenic lamin A/C malignant VAs with 2 risk factors may benefit from
22281253 lamin A/C mutation carriers mutation carriers prophylactic ICD
between 2000 and Results:
Study type: Cohort, 2010 48 (18%) had malignant VAs (11
multicenter successful CPR, 25 appropriate
Exclusion criteria: ICD treatment, and 12 died
Size: 269 patients Patients 15 y of age suddenly)
Median follow up of Risk factors for VAs were NSVT,
43 mo LVEF <45%, male sex, and non-
missense mutations (ins-
del/truncating or mutations
affecting splicing). VA occurred
only in persons with at least 2 of
these risk factors.
Meune et al. Aim: To assess whether ICD is Inclusion criteria: 1 endpoint: Not specified 1 inappropriate shock
2006 (319) beneficial for 1 prevention of Lamin A/C mutations Summary: ICD rather than pacemaker
16407522 SCD in patients with lamin A/C associated with cardiac Results: should be considered in patients with
gene mutations with conduction defects 8/19 (42%) received conduction disorders and lamin A/C
preserved LVEF referred for appropriate ICD therapy mutation
pacing due to presence of Exclusion criteria: Follow up 33.921 mo
progressive conduction delay 19 patients received No factor (including LVEF,
or SND ICD (Muscular spontaneous or induced VA or
phenotype: 9 Emery- drug therapy) predicted VA
Study type: Cohort study Dreifuss, 8 DCM plus events
conduction disease, 1 LVEF not reduced in patients
Size: 19 patients Limb-girdle, 1 receiving ICD therapies
shoulder-muscle
amyotrophy)
Mean age 41.713.4
y
Sex: 73% Male
Mean LVEF 58%12%
Pasotti et al. Aim: The aim of this study was Inclusion criteria: 27 1 endpoint: Events were death Authors concluded that dilated
2008 (210) to analyze the long-term consecutive families in from any cause, death from HF, cardiomyopathies caused by LMNA
18926329 follow-up of dilated which LMNA gene heart transplantation, and SCD, gene defects are highly penetrant,
cardiolaminopathies in defects were identified adult onset, malignant diseases

177

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
patients with Lamin A/C gene in the probands, all including appropriate ICD characterized by a high rate of HF and
mutations sharing the DCM interventions life-threatening arrhythmias.
phenotype. Of the 164
Study type: Retrospective family members, 94 Results:
observational longitudinal had LMNA gene 60 of 94 (64%) were
study mutations phenotypically affected whereas
34 were only genotypically
Size: 94 patients Exclusion criteria: N/A affected.
Of the 60 patients, 40 had DCM
with AVB, 12 had DCM with
VT/fibrillation, 6 had DCM with
AVB and EDMD2, and 2 had AVB
plus EDMD2.
During a median of 57 mo there
were 49 events in 43 DCM
patients.
The events were related to HF
(15 heart transplants, 1 death
from end-stage HF) and VA (15
SCDs and 12 appropriate ICD
interventions).
van Berlo et Aim: To evaluate common Inclusion criteria: 21 1 endpoint: Arrhythmias and Authors conclude that carriers of
al. 2005 (211) clinical characteristics of publications between sudden death lamin A/C mutations carry a high risk of
15551023 patients with lamin A/C gene March sudden death.
mutations that cause either 1999 and March 2002 Results: Presence of pacemaker did not
isolated DCM or DCM in reporting lamin A/C Cardiac dysrhythmias were protect against sudden death.
association with skeletal gene mutations reported in 92% of patients after
muscular dystrophy. 30 y of age; HF was reported in
Exclusion criteria: 64% after 50 y of age.
Study type: Meta-analysis Patients with familial 76 of the reported 299 patients
(pooled data) partial lipodystrophy, (25%) died at a mean of 46 y of
progeria, axonal age.
Size: 299 carriers of neuropathy and Sudden death was the most
lamin A/C mutations mandibuloacral frequently reported mode of
dysplasia caused by death (46%) in both the cardiac
mutations in the lamin and the neuromuscular
phenotype.

178

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
A/C gene were
excluded
Lallemand et Aim: To analyze the natural Hx Inclusion criteria: 1 endpoint: N/A In patients with normal initial EPS,
al. 2012 (320) and predictors of change in Patients with muscular changes in the resting ECG and/or SA-
22038543 infra-Hisian conduction time dystrophy of which 25 Results: Mean HV interval ECG on annual follow-up were
in myotonic dystrophy underwent a second increased between the baseline associated with change in infra-Hisian
patients with normal baseline EPS for new symptoms, and follow-up EP conduction
EPS new AV conduction Study 52.11.6 ms to 61.42.2
abnormalities on ECG, ms.
Study type: Cohort study changes on SA-ECG, Predictors of increased HV
ord asymptomatic interval were change in resting
Size: 127 patients patients >60 mo from ECG and SA-ECG (QRSd 100 ms
first EPS or low amplitude signal <40
microvolts)
Exclusion criteria: N/A 5 patients with HV 70 ms
received prophylactic pacemaker
Wahbi et al. Aim: To determine whether Inclusion criteria: 1 endpoint: All-cause mortality In patients with myotonic dystrophy
2012 (321) an invasive strategy based on Genetically confirmed type 1, an invasive strategy was
22453570 EPS and prophylactic myotonic dystrophy Results: associated with a higher rate of 9y
pacemaker is associated with type 1 with PR >200 ms 341 (70.2%) - EPS survival than a noninvasive strategy
longer survival in patients and/or QRS >100 ms compared to 145 (29.8%) -
presenting with myotonic between 1/2000 to noninvasive strategy
dystrophy type 1 and 12/2009
infranodal conduction delays Median follow-up 7.4 y (322)
compared to a noninvasive Exclusion criteria: N/A 50 patients died in EPS strategy
strategy using propensity group
adjustments 30 died in the noninvasive
strategy group (HR: 0.74; 95% CI:
Study type: Cohort study 0.471.16; p=0.19)
Difference attributable to a
Size: 486 patients lower incidence of SCD (10
patients invasive strategy group
vs. 16 patients noninvasive
strategy group, HR: 0.24; 95% CI:
0.100.56; p=0.001])

179

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Ha et al. 2012 Aim: To define predictors of Inclusion criteria: 1 endpoint: N/A Despite identification of conduction
(323) cardiac conduction disease in Patients with DM1 and disease and prophylactic pacing,
22385162 myotonic dystrophy patients 25 DM2 after 2003 Results: mortality remains high in patients with
Follow-up 5746 mo a severe ECG abnormality (most deaths
Study type: Cohort study, Exclusion criteria: N/A non-sudden, suggesting that a severe
single-center A severe ECG abnormality was ECG abnormality is also general marker
defined as a PR interval of 240 of risk for all-cause mortality.)
Size: 211 patients ms or QRS duration of 120 ms Of 3 patients who died suddenly, 2
had pacemakers, suggesting that a
Severe ECG abnormality severe ECG abnormality does not
present in 24% of DM1 patients simply predict sudden death from AV
and 17% of DM2 patients block
Pacemaker or ICD implanted in
14% of all patients, including 65%
of patients with severe ECG
abnormalities.
13 patients died (1.16%/y),
including 3 sudden (2 of whom
had pacemakers)
Laurent et al. Aim: To determine whether Inclusion criteria: 1 endpoint: All-cause mortality Implantation of a pacemaker when
2011(324) implantation of prophylactic Genetically confirmed HV interval 70 seemed to identify a
20227121 pacemaker in myotonic MD1 between 1994 Results: population likely to progress to high
dystrophy patients with HV and 2008 at single 10 deaths (9 respiratory failure, grade AV block. A higher rate of
interval 70 lowers the risk of institution 1 sudden). 1 SCD occurred in a sudden death would have been
sudden death (due to patient with pacemaker who had expected based on previous studies of
complete AV block) Exclusion criteria: no spontaneous VT suggesting a comparable populations, implying that
Infantile form of MD non-cardiac etiology for this prophylactic pacemaker implantation,
Study type: Cohort study 100 patients enrolled event. based on these criteria, may have
and 49 implanted with 1/51 with HV interval <70 prevented some deaths due to
Size: 100 patients pacemaker for HV developed complete AV block asystole.
interval 70 19/49 patients with HV 70
Mean follow up developed AV block
7439 mo
46% had 1 or more
Groh criteria (rhythm
other than sinus, PR
240 ms, QRS 120 ms,

180

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
2nd or 3rd degree AV
block)
Bhakta et al. Aim: To assess implant rates Inclusion criteria: 1 endpoint: N/A Adult DM1 patients commonly
2011 (325) and indications for pacemaker Genetically confirmed receive pacemakers and ICDs.
22035077 and ICDs and outcomes in DM1 Results: The risk of SCD in patients with
patients with DM1 Follow up 9.53.2 y pacemakers suggests that the ICD may
Exclusion criteria: N/A 46 (11.3%) received a pacemaker warranted but SCD was still observed in
Study type: Cohort study, and 21 (5.2%) an ICD ICD patients raising uncertainty benefit.
multicenter Devices were primarily implanted DM1 patients are at high risk of
for asymptomatic conduction respiratory failure. Therefore,
Size: 406 patients abnormalities or LV systolic pacemaker or ICDs in asymptomatic
dysfunction patients moderate conduction disease
and also severe skeletal muscle
7 (15.2%) pacemakers were involvement may not improve
implanted for third-degree AV outcomes.
block and 6 (28.6%) ICDs were
implanted for VAs
5 (10.9%) pacemaker patients
underwent upgrade to an ICD (3
for LV systolic dysfunction, 1 for
VAs, and 1 for progressive
conduction disease).
17 (27.4%) of the 62 patients
with devices were pacemaker-
dependent at last follow-up
3 (14.3%) ICD patients had
appropriate therapies
24 (52.2%) pacemaker patients
died including 13 of respiratory
failure and 7 of sudden death
7 (33.3%) ICD patients died
including 2 of respiratory failure
and 3 of sudden death (1 death
was documented due to
inappropriate therapies)

181

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Nazarian et al. Aim: To characterize the Inclusion criteria: 1 endpoint: Time dependent PR Patients with DM1 can develop rapid
2011 (326) trends and predictors of time- Patients with DM1 or QRS prolongation during changes in cardiac conduction intervals.
20946286 dependent ECG changes in baseline ECG and then follow-up AF or flutter, older age, and larger
patients with DM1 routine follow-up CTG expansions predict greater time-
Results: dependent PR and QRS interval
Study type: Cohort study, Exclusion criteria: Age, h/o AF or flutter, and prolongation and warrant particular
single center History of second or number of cytosine-thymine- attention in the arrhythmic evaluation
third degree AV block, guanine (CTG) repeats were of this high-risk patient subset.
Size: 70 patients VAs, resuscitated SCD, predictors of time-dependent PR
or persistent supraVA and QRS prolongation
Mean follow-up 956 Lower LVEF associated greater
d QRS progression
Clinical predictors of
conduction disease
progression were
assessed using
multivariate analysis
Bhakta et al. Aim: To assess the prevalence Inclusion criteria: 1 endpoint: N/A There is a notable incidence of LV
2010 (327) of conduction disease and Patients with DM1 with systolic dysfunction and HF exists in
21146669 LVEF in population of patients confirmed abnormal Results: patients with DM1.
with DM1 CTG repeat sequence Cardiac imaging was performed The presence of LVSD/HF in DM1 is
(one or both alleles on 180 (44.3%) significantly associated with all-cause
Study type: cohort study, 38 repeats) and cardiac death.
multicenter Prevalence of LV systolic
Exclusion criteria: dysfunction and HF in 41 (10.1%)
Size: 406 patients Patients <18 y or of 406 (risk factors were
unconfirmed DM1 increasing age, male sex, ECG
diagnosis as above conduction abnormalities,
presence of atrial and VA, and
implanted devices)
Presence of decreased LVEF
was associated with all-cause
death (RR: 3.9; 95% CI: 2.36.4;
p<0.001) and cardiac death (RR:
5.7; 95% CI: 2.612.4; p<0.001).

182

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Groh et al. Aim: To identify whether the Inclusion criteria: 1 endpoint: N/A Patients with DM1 are at high risk for
2008 (328) ECG is useful for prediction of Genetically confirmed sudden death (up to 1/3 of deaths are
18565861 SCD risk in patients with DM1 DM1 (only patients Results: sudden)
with abnormal CTG Defined: Severe abnormality on Severe abnormality on ECG (RR: 3.3;
Study type: Cohort study, repeat sequence 38 ECG includes rhythm other than 95% CI: 1.258.78) and diagnosis of
multicenter repeats) sinus, PR interval 240 ms, QRS atrial tachyarrhythmia (RR: 5.18; 95%
120 ms, or 2nd or 3rd degree AV CI: 2.2811.77) predictive of sudden
Size: 406 patients Exclusion criteria: N/A block death in patients with DM1
96/406 had severe abnormality Severe abnormality on ECG PPV
on ECG 9 received ICD and 23 12.1% and NPV 97.1% for prediction of
pacemakers SCD

Follow-up 5.7 y during which


81/406 (20%) died (27 SCD, 32
respiratory failure, 5 non-sudden
cardiac deaths, 17 deaths from
other causes)
Of the 27 SCD, 17 had post-
collapse rhythm documented of
which only 9 was VT/VF
Severe abnormality on ECG (RR:
3.3; CI: 1.258.78) and diagnosis
of atrial tachyarrhythmia (RR:
5.18; CI: 2.2811.77) predictive of
sudden death in patients with
DM1
Rates of prophylactic pacing
increased during the study period
and we not associated with
decreased rates of SCD
Lafort P et al. Aim: Evaluate the incidence of Inclusion criteria: 1 endpoint: N/A Patients with FSHMD may have
1998 (329) cardiac involvement in Patients exhibiting cardiac involvement.
9818880 facioscapulohumeral muscular clinical and molecular Results: 5 patients had Significant clinical cardiac
dystrophy features of conduction defects or arrhythmia involvement is rather rare in this form
facioscapulohumeral (IVCD or AF/flutter induced by of muscular dystrophy, specific
Study type: Cohort, single muscular dystrophy EPS), 1 case of AV block requiring monitoring or treatment
center recommendations are not well defined.

183

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: N/A pacemaker, 1 case of VT possibly Discussion of arrhythmia- related
Size: 100 patients related to co-existing ARVC symptoms and yearly
electrocardiograms has been
recommended.
Stevenson et Aim: Evaluate incidence of Inclusion criteria: 1 endpoint: Evidence of cardiac Evidence supporting cardiac
al. 1990 (330) cardiac involvement in Patients with involvement involvement in this condition with
2299071 fascioscapulohumeral fascioscapulohumeral minority of cases having abnormal
muscular dystrophy muscular dystrophy Results: sinus node function or AV conduction.
(autosomal dominant 30/30 had 12-lead ECG, 22/30
Study type: cohort, single inheritance, had 24 hr Holter, 15 had
center characteristic facial echocardiogram, 10 patients had
involvement, 12 EP studies
Size: 30 patients scapular/deltoid
muscle weakness > P wave abnormalities were
biceps/triceps, common (60%)
myopathic changes on AF or Aflutter induced at EPS in
biopsy or EMG) 10/12
Evidence of abnormal AV node
Exclusion criteria: conduction or infranodal
Elbow contractures, conduction present on EPS or
absence of scapular ECG in 27% of patients
winging, and X-linked Sinus node function abnormal
heredity in 3 patients

Data Supplement 39. Nonrandomized Trials Related to Cardiac Channelopathies (Section 7.9)
Study Acronym; Study 1 Endpoint and Results
Summary/Conclusion
Author; Type/Design; Patient Population (P values; OR or RR;
Comment(s)
Year Published Study Size & 95% CI)
Costa J et al. HR Study type: Inclusion criteria: LQT1 1 endpoint: LQT1 gender and mutation Combined assessment of clinical
2012 (331) multicenter gentoype, age 0-40 y specific risk stratification ACA/SCD and mutation location can identify
22293141 gender specific risk factors for life-
Size: 1051 Exclusion criteria: Results: Increased risk: threatening events
Age 0-13 y: males; >13, Males =females
Loop mutations: HR: 2.7 for females, not
males

184

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Time-dependent syncope increased risk for
males, HR: 4.73
QTc 500 ms: higher risk for women

Bai R, et al. Study type: Inclusion criteria: 1 endpoint: Yield of genetic testing and Yield in LQTS higher if confirmed
CAE 2009 (332) Sigle center consecutive probands cost dx present: 64%
19808439 retrospective referred with confirmed or Yield in BrS increased if type 1 BrS
suspected LQTS, BrS, or Results: Yield and cost in US $ per diagnosis: ECG with AV block present
Size: 1394 CPVT, or idiopathic VF/ACA LQTS: 40%, $13402 Yield in CPVT increased in males,
Br S: 8%, $33,148 prior CA, or confirmed bidirectional
Exclusion criteria: N/A CPVT: 35%, $9170 VT present
Idiopathic VF: 9%, $71,430 LQTS, CPVT reasonable cost if
strong clinical suspicion
BrS less cost effective
Idiopathic VF ineffective, costly
Gehi AK, et al. Study type: Inclusion: Publications 1 endpoint: Identify risk predictors of BrS ACE risk increased with prior
JCE 2006 (333) Meta-analysis: 1/1990-3/2005 on adverse natural history in patients with syncope or SCD, RR: 3.24
16836701 retrieved 30 prognosis of patients with a Brugada ECG Males, RR: 3.47
prospective Brugada ECG: Spont type 1 ECG RR: 4.65
studies on Prospective cohort studies, Results:
Brugada ECG >10 subjects, primary data Risk increased with prior hx syncope or ACA,
on syncope, SCD, ICD spont type 1 Br ECG, and male gender
Size: 1545 shocks; followup >6 mo and
>90% followup NOT sig risk factors: Fam hx SCD
SCN5A mutation, or inducibility by PES: (not
Exclusions: non-English; a risk factor but heterogeneity of studies)
presence of cardiac disease
Kim JA et al. HR Study type: Inclusion criteria: genotype 1 endpoint: LQT2 genotype: trigger Pore-loop mutations assoc with
2010 (334) multicenter + LQT2 specific risk factors for SCD/ACA arousal events;
20850565 retrospective BB not significanty protective for
Exclusion criteria: N/A Results: arousal 44%, exercise 13%, non- this subset
Size: 634 exercie/non-arousal 43%
Risk for arousal: female >13 y, pore-loop
mutation

185

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Non-pore loop assoc with exercise events,
HR:6.84
Beta-bl reduced risk for exercise events but
not arousal/non-exercise events
Migdalovich D Study type: Inclusion criteria: 1 endpoint: LQT2 genotype vs outcome Women w LQT2 much higher risk v
et al. HR 2011 multicenter LQT2 genotype ACA/SCD by age 40 y men
(335) retrospective Pore-loop vs non-pore loop mutations Overall, pore loop mutations sig
21440677 Exclusion criteria: N/A increased risk ACA, SCD, greater risk
Size: 1166 Results: women w LQT2 much higher risk: for males vs females
26% vs. men; Pore loop mutations LQT2 males,
For women, no sig difference in mutation HR:2.18 for ACA/SCD
site
Risk similar at age <13 y;
Age >13 y, females HR: 2.23 ACA/SCD vs
males
Males: pore loop mutations >2-fold
increased risk
Increased risk: QTc 500 msec (males 2x,
females 4-fold increase)
Highest risk: 5.3/1000 patient-y: prior
syncope plus QTc 500 ms, pore loop male,
or female >13 y old, HR: 17
BB: 61% reduced risk
Ackerman MJ Study type: Expert consensus statement General: Class I: 1) sound clinical suspicion LQTS: Note difference between
2011 (182) HRS/EHRA on the state of genetic when positive predictive value > 40%, Class I if QTc >480 or 500 ms, and
21810866 consensus testing for the signal/noise ratio >10; 2) AND/OR genetic Class IIb if QTc > 460/480 ms
statement. channelopathies and test result provides either diagnostic or
cardiomyopathies prognostic info, or influences therapeutic
choices.
Panel: geneticists, Screening of family members: when genetic
arrhythmia specialists testing leads to the adoption of
Agreement 84% therapy/protective measures/ lifestyle
adaptations.

LQTS: Class I: 1) any pt with strong clinical


index of suspicion for LQTS; 2) any
asymptomatic pt with QT prolongation on

186

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
serial ECGs: QTc >480 ms prepuberty; >500
ms, adult; 3) Mutation specific genetic
testing for family members and other
appropriate relatives
Class IIb: any asymptomatic pt with
otherwise idiopathic QTc values >460 ms
(puberty) or 480 ms on serial ECGs

CPVT: Class I: 1) any pt w strong clinical


index of suspicion of CPVT;
2) Mutation specific genetic testing is
recommended for family members and
appropriate relatives

Brugada: Class I: Mutation specific genetic


testing is recommended for family members
and appropriate relatives
Class IIa: any pt w strong clinical index of
suspicion of BrS, including with
procainamide challenge
Class III: not indicated in the setting of an
isolated type 2 or 3 Brugada ECG pattern

Short QTS: Class I: Mutation specific genetic


testing is recommended for family members
and appropriate relatives
Class IIb: any pt with strong clinical index of
suspicion

ARVC: Class I: Mutation specific genetic


testing is recommended for family members
and appropriate relatives
Class IIa: can be useful for patients satisfying
task force diagnostic criteria
Class IIb: may be considered for patients
with possible ACM/ARVC

187

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Class III: not recommended for patients with
only a single minor criterion according to
the 2010 task force criteria

SCD/SIDS: Class I: 1) Collection of tissue


sample recommended (blood or
heart/liver/spleen tissue); 2) Mutation
specific genetic testing is recommended for
family members and appropriate relatives
Class IIb: testing may be considered if
circumstantial evidence suggests LQTS or
CPVT specifically

ACA/resuscitated: Class I: Genetic testing


should be guided by the results of medical
evaluation and is used for the 1 purpose of
screening at-risk family members for sub-
clinical disease
Class III: Routine genetic testing, in the
absence of a clinical index of suspicion for a
specific cardiomyopathy or channelopathy,
is not indicated for the survivor of
unexplained OHCA

HCM: Class I: 1) any pt in whom the clinical


dx of HCM is established. 2) Mutation
specific genetic testing is recommended for
family members and appropriate relatives

DCM: Class I: 1) DCM and significant cardiac


conduction disease and/or family Hx of
premature unexpected sudden death. 2)
Mutation specific genetic testing is
recommended for family members and
appropriate relatives

188

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
LVNC: Class I: Mutation specific genetic
testing is recommended for family members
and appropriate relatives
Class IIa: can be useful if clinical dx of LVNC
is established

PCCD: Class I: Mutation specific genetic


testing is recommended for family members
and appropriate relatives
Class IIb: may be considered as part of
diagnostic evaluation for patients with
either isolated CCD or CCD with
concomitant congenital heart disease,
especially w post family Hx of CCD.

Nannenberg EA Study type: Inclusion criteria: 1 endpoint: Using FTMR method to Identify age ranges of highest risk
Circ CV Genetics Retrospective Genotype positive 6 achieve Standardized Mortality Ratio(SMR) for specified inherited arrhythmia
2012 (336) single center, inherited arrhythmia (observed to expected mortality by syndromes
22373669 Netherlands syndromes analyzed with genotype and age in inherited arrhythmias Asymptomatic patients over age
Family Tree Mortality Ratio ranges may not require rx
Size: 1170 (FTMR): Results: LQTS1: in first 10 y of life SMR 2.9
LQT1,2,3; Brugada (1.55.1)
Syndrome, SCN5A overlap LQTS2: age 30-39 y, SMR 4.0 (1.110)
syndrome (LQT3, BrS, LQTS3: age 15-19 y, SMR 5.8(1.216.9)
conduction disease); RYR2 SCN5A overlap syndrome: 20-39 y, SMR 3.8
CPVT. (2.55.7)
CPVT: age 20-39 y, SMR 3.0 (1.36.0)
Exclusion criteria: N/A BrS: 40-59 y, SMR 1.79 (1.22.4), especially
males
Kimbrough J Study type: Inclusion criteria: 791 first 1 endpoint: Risk of ACE for family Affected female parents have
Circ 2001 (337) Retrospective degree relatives of 211 LQTS members of proband with LQTS increased risk of cardiac event
11479253 multi-center probands before age 40 y.
Results: Severity of proband symptoms did Severity of proband symptoms did
Size: 791 Exclusion criteria: N/A not significantly influence family members not significantly influence family
symptoms, although more likely to receive members symptoms.
BB.

189

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Female gender and duration of QTc
important risk factors

Kaufman ES Study type: Inclusion criteria: Patients 1 endpoint: risk of death in LQTS when a SCD of sibling did not predict risk
Heart Rhythm Retrospective with QTc 450 msec in sibling has died: ACA, SCD, or syncope of death or ACA
2008 (338) registry: registry, who had a sibling Did correlate with increased risk of
18534367 International with SCD Results: 270 patients with sibling SCD syncope ~6%
LQTS Registry Sibling death did not correlate with risk Hx of syncope, QTc 530 msec,
Exclusion criteria: N/A ACA/SCD female gender correlated with
Size: 1915 Was associated with increased risk of increased risk ACA/SCD
syncope
Associations with increased risk death: QTc
530 msec, syncope, gender
Wedekind H Eur Study type: Inclusion criteria: 1 endpoint: Recurrent syncope, ACA or Risk predictors: QTc > 500 msec,
J Ped 2009 (339) Retrospective Genotype positive SCD after dx LQTS. Mean followup 5.94.7 y prior syncope or ACA
19101729 single center probands, age 16 y LQTS: LQT2 highest rate SCD vs other
89% LQT1, 2,3 Results: 92% treated: Followup:
Size: 83 Mean QTc 51074 ms Propranolol 79%, atenolol 20%, metoprolol
61% symptoms: syncope 12%, bisoprolol 8%, pindolol 2%; mexiletine
49%, ACA 33%, SCD 18% 4%
78% with BB rx ICD 8%, pacer 5%.
31% recurrent symptoms: 14% ACA or SCD;
Exclusion criteria: N/A syncope 86%
Significant predictors: QTc >500 ms (HR: 2.9;
95% CI: 1.27.3 p=0.02); prior syncope HR:
4.04; 95% CI: 1.115, ACA HR:11.7; 95% CI:
3.143.4, p<0.001
Goldenberg I Study type: Inclusion criteria: 1 endpoint: LQTS with normal QTc: risk for Genotype positive patients with
JACC 2011 (340) Multicenter Genotyped patients with ACE: ACA or SCD normal QTc =25% of genotype
21185501 international LQTS: 3386 patients positive patients.
registry, Normal QTc: 440 ms Results: Normal QTc =14% of total LQTS 4% ACA/SCD with normal QTc vs
retrospective Prolonged QTc >440 ms patients in study. 15% if prolonged QTc
Unaffected: negative Normal QTc risk ACA/SCD =4%, lower than
Size: 469 genotype those with prolonged QTc (15%) but higher
than genotype neg family members.
190

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: N/A Increased risk: mutation characteristics;
LQT1 vs LQTS 2, HR: 9.88; p=0.03;
Duration of QTc and gender important only
in those with prolonged QTc.

Tester DJ JACC Study type: Inclusion criteria: 1 endpoint: yield of LQTS genetic testing Genotype results more likely to be
2006 (341) retrospective consecutive patients vs. clinical genotype positive with QTc >480ms or with
16487842 single center undergoing Genetic testing higher Schwartz score
LQTS 1997-2004 Results: 50% positive genotype. Yield
Size: 541 correlated with duration of QTc and
Exclusion criteria: N/A phenotype: 0%: QTc<400
62%: QTc >480 ms (p<0.0001)
Schwartz score 4: 72% positive
Priori S Circ Study type: Inclusion criteria: Brugada 1 endpoint: Brugada risk stratification for Multivariable risk predictor:
2002 (342) Multicenter S with ECG changes, spont SCD spontaneous ST elevation V1-V3 and
11901046 retrospective (51%) or induced PES performed in 86 Hx of syncope
130 probands Syncope without spontaneous ST
Size: 200 Results: SCN5A identified in 22% probands, elevation not a risk factor
Exclusion criteria: N/A 46% of family members PES not predictive
Risk analysis: gender; ECG, family hx, Mutation carriers without
mutation status, symptoms phenotype: low risk
Syncope without ST elevation on baseline
ECG: not a risk
Syncope AND ST elevation: increased risk
SCD, HR: 6.4; p <0.002
FINGER Study type: Inclusion criteria: Brugada 1 endpoint: ACE outcomes in BrS Low event rate in asymptomatic
Probst V Circ Multi-center Syndrome patients 0.5%/y.
2010 (343) registry, 11 ECG spont (45%) or with Results: PES performed in 62%: 41% Inducibility w PES or family Hx SCD
20100972 centers in drug challenge. positive, higher in symptomatic patients or SCN5A mutation not predictors of
Europe Median 45 y (35-55). 46% vs 37%, p=0.02. ACE
Hx ACA 6%, syncope 30%, PES performed in 369 asymptomatic Predictors of ACE: symptoms, ACA,
Size: 1029 asymptomatic 64% (654 patients: 37% positive (137/369); 85% syncope, presence of ICD, spont type
patients). (117/137) inducible asyx patients had ICD 1 ECG.
SCN5A positive 22%. implanted Among asymptomatic patients:
ICDs implanted: 433/1029 patients (42%): 37% positive PES; of these 85% had
Exclusion criteria: N/A of 433: 54 ACA (12.5%), 208 syncope (48%), ICD implanted.

191

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
171 asymptomatic (39%). 118/171 ICD implantation in asymptomatic
asymptomatic patients with ICD (69%) patients was significant in
implanted due to positive EPS. multivariable analysis as predictor of
ACE: HR:10.1; 95% CI: 1.758.7,
ACE 51: approp ICD shocks 44, SCD 7. p=0.01).
Mean ACE rate 1.6%/y: 7.7% in patients w No independent predictive value of
Hx ACA;1.9% w prior syncope; 0.5% in PES (p=0.09), males (p=0.42, spont
asymp patients type 1 ECG (p=0.38) age (p=0.97)
Predictors: symptoms (p<0.001): ACA (HR:
11; 95% CI: 4.824.3, p<0.001), syncope (HR:
3.4; 95% CI 1.67.4, p=0.002),
ICD implantation (HR: 3.9; 95% CI: 1.410.6,
p=0.007).
spont type 1 ECG (HR: 1.8;95% CI: 1.03
3.33, p=0.04);
NOT predictive: gender, family Hx SCD, +PES
(p=0.48), presence SCN5A mutation
Moss AJ Circ Study type: Inclusion criteria: LQTS 1 endpoint: Recurrent CE on b-bl in LQTS For LQT 1 and 2, BB reduce risk
2000(344) Retrospective registry, Rochester, patients Highly symptomatic patients
10673253 observational treatment w BB age <41 y, Results: B-Bl significantly reduce risk LQT 1 prior to treatment at high risk
80% syncope or ACA prior and 2; for recurrent events.
Size: 869 to rx. Atenolol, metoprolol, LQT 3: no effect LQT 3 patients: BB did not
nadolol, propranolol. For symptomatic patients, HR 5.8 for reduce risk
139/869 genotyped: LQT recurrent CE: 32% ACE within 5 y.
1(69), LQT 2 (42), LQT 3 (28) Prior syncope: HR: 3.1.
Exclusion criteria: age >41 y Prior ACA, HR: 12.9 for ACA or sudden
start rx death: 14% recurrent CA.
Zareba JCE 2003 Study type: Inclusion criteria: 125 LQTS 1 endpoint: Mortality of LQTS patients Prior ACA or recurrent syncope on
(345) Single center patients with ICDs treated with/without ICD: b-bl treatment assoc with significant
12741701 retrospective compared with LQTS with 73 patients with syncope on treatment or mortality without ICD during 8 y
similar risk and no ICD. ICD prior ACA and ICD compared with 161 LQTS followup
Size:125 Indications: 54 ACA, 19 patients without ICD (89 ACA, 72 rec
recurrent syncope on b-bl; syncope on b-bl)
52 other (syncope; +
family Hx SCD) Results: Deaths: ICD 1.3% (1 pt), followup
av 3 y, vs. 16% (26 patients) in non-ICD
Exclusion criteria: N/A patients during 8 y mean followup.

192

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Monnig G Heart Study type: Inclusion criteria: 1 endpoint: LQTS Appropriate ICD shocks Predictors of approp ICD shocks:
Rhythm 2005 single center symptomatic LQTS patients or death during followup. QTc >500 msec, prior ACA
(346) retrospective undergoing ICD implant. Approp shocks reduced by anti-
15840474 Mean QTc 54064; 85% Results: Mean followup 6534 mo. brady pacing, b-bl rx, rate-smoothing
Size: 27 famle, 63% ACA, 33% Death 1 pt, non-cardiac.
recurrent syncope on b-bl, Approp shocks: 37%; 30% multiple shocks.
4% severe phenotype Logistic regression: QTc >500 ms, prior ACA
81 genotype pos: LQT 1 28, predictive.
LQT2 39; LQT3 1, LQT5 13. Shocks reduced from av 7.1 to 0.75 shocks
annually by adding b-bl, increased rate anti-
Exclusion criteria: N/A brady pacing, rate smoothing algorithm.
Hayashi M Circ Study type: Inclusion criteria: CPVT 50 1 endpoint: ACE in CPVT patients: Higher risk for lack of BB, Hx ACA
2009 (347) single center probands, 51 family syncope, ACA, approp ICD shocks, SCD Prior syncope not associated with
19398665 retrospective members, age at dx 1510 increased risk
y. Results: followup 7.9 y
Size: 101 Symptoms 60% (61 8 y total event rate 32% total, 27% with b-bl,
patients), all probands, 22% 58% without b-bl. 8 y event ACA/SCD 13% (8
family members patients)
93% symptomatic <21 y old Increased risk: Absence BB HR: 5.54; 95% CI:
77% detection of mutations: 1.1716.15, p=0.003), Hx ACA HR: 13.01;
RYR2 CASQ2 95% CI: 2.4868.21, p=0.002); younger age
at dx (HR: 0.54/decade; 95% CI: 0.330.89,
Exclusion criteria: N/A p=0.02)
32% with events on b-blockers did not take
meds on day of event.
Nadolol: ACE 19%
Delise P EHJ Study type: Inclusion criteria: Type 1 1 endpoint: predictors in Brugada S of ACE Combining 2 or more risk factors
2011(348) Multi- center Brugada ECG: spontaneous (approp ICD shocks, sudden death) was useful risk stratification:
20978016 prospective 54%, drug-induced 46%. Spontaneous type 1 ECG
Results: Median followup 40 mos (IQR 20- Family Hx sudden death,
Size: 320 Median age 43 y. 67) syncope, positive PES
Males 81% 5.3 % MACE (17 patients): VF on ICD (14),
sudden death3 MACE occurred only in patients
Asymptomatic 66%, MACE occurred in 10.4% of symptomatic with 2 or more risk factors. MACE
syncope 33% and 2.8% of asymptomatic patients event rates:
(p=0.004) 3.0%/pt/yr in symptomatic,
NO prior ACA ICDs implanted in 34%(110 patients) 0.8%/pt/yr in asymptomatic

193

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
PES performed in 245 (76%): positive in 50%
of symptomatic and 32% of asymptomatic PES can be useful in patients with
Exclusion criteria: N/A patients. spontaneous type 1 ECG and no
MACE in 14% of positive PES, 0% of other risk factors; may be helpful to
negative, 5.3% of no EPS: positive predictive identify low risk patients
values 14%, negative pred value 100%
VF occurred in 15.5% of patients with
inducible VF using doubles, 8.6% of triples
Combination of risk factors most significant:
spont ECG, family Hx sudden death,
syncope, positive EPS: no events occurred in
patients without any of above or with only
one risk factor.
Spontaneous type 1 ECG: if additional risk
factors, 30% MACE (p<0.001)
Hiraoka M JE Study type: Inclusion criteria: Brugada 1 endpoint: Brugada S ages 18-35 y at dx, Brugada outcomes in young adults
2013 (349) Prospective S patients ages 1835 y outcomes of VF or SCD vs presenting symptoms:
23702150 single center Mean age 306 y Followup 4327 mos. Events: VF 11.2% /y, syncope 3.3%
y, asymptomatic 0.7%/y
Size: 69 No genetic testing Results: Based on presenting symptoms:
VF 42%, syncope 12%, asymptomatic 2.5%
Not predictive: gender, family Hx SCD, abnl
Exclusion criteria: N/A SAECG, spontaneous vs drug-induced ECG,
inducible VT/VF

All ages 460 patients symptoms at


presentation vs outcomes:
VF 8.4%/y, Syncope 1.7%/y, asymptomatic
0.3%/y

PRELUDE Study type: Inclusion criteria: Age >18 1 endpoint: Predictive accuracy of PES for PES did not predict high risk
Priori SG et al. Prospective y, BrS type 1 ECG spont sustained VT/VF or approp ICD shock in Predictors: spontaneous type BrS
JACC 2012 registry (56%, 171/308) or drug- Brugada. ECG and symptoms; f-QRS, VERP
22192666 induced, without prior ACA; <200 msec
Size: 308 Results: PES performed at enrollment; VERP <200 msec was predictive: this
followup every 6 mo. data would only be obtained at EPS.
Mean age 4512 y.

194

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
21% with prior syncope (65 Cardiac arrest 4.5% (14/308), 13/14 NOTE that + PES used in decision
patients: 16/65 {25%} > 1 resuscitated with ICD, EMS 1. to implant ICDs: 13/137 patients
syncope). PES positive in 41% (126/308); of these: (9.5%) with ICDs were resuscitated
single stimulation 5.5%, double 44.5%, with ICD.
SCN5A positive 20% of triples 50%.
tested patients. ICDs implanted in 137 patients (78% of Note 1/14 patients with VF had only
inducible patients {98/126} and 21% of non- spont type 1 ECG and no prior
(f-QRS =2 or more spikes inducible patients {39/182}. syncope, neg family hx, neg EPS,
within QRS leads V1-V3: Annual event rate 1.5%: VERP >200 msec but + SCN5A
present 8.1%) mutation and received ICD after EPS.
Multivariable predictors: spont type 1 ECG Only 1 pt without ICD had ACA: pt
Exclusion criteria: N/A and Hx of syncope (HR: 4.20; 95% CI: 1.38 had spont type 1 ECG, VRP <200
12.79, p=0.012), Ventricular ERP <200 msec msec, and fQRS.
(HR: 3.91; 95% CI: 1.0312.79, p=0.045),
QRS fractionation (HR: 4.94, 95% CI: 1.54
15.8, p=0.007).

Positive PES not predictive (HR: 1.03; 95%


CI: 0.343.16, p=0.96)
Wilde A et al. Study type: Inclusion criteria: LQT3 1 endpoint: LQT3 ACE outcomes: syncope, High risk LQT3:
Circ 2016 multicenter SCN5A mutation carriers ACA, SCD Females;
27566755 observational Median followup 7 y syncope, QTc 450-490
In 8%, first cardiac
Size: 391 symptom: ACA, SCD Results: Rx: B-bl 29%; LCSD 2%; pacer 5%; Hx of syncopedoubled risk
ICD 18%.
Exclusion criteria: Time dependent increase in ACE: by age BB therapy significantly reduced
symptoms during first year 40yrs, ~40% with ACE. ~ 50% of ACE =ACA or risk for ACE, especially in females
of life-12 patients; SCD
Lost to followup after age 1: Mutation type/location did not have
3 patients; B-blocker rx: 83% risk reduction in females sig effect on outcome
Patients with 2 mutations (p=0.015); 49% risk reduction in males (not
sig; too few events in males to assess)
BB not pro-arrhythmic
3% died on BB during followup
Multivariate risk factors: QTc, syncope:

195

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Each 10 msec increase in QTc up to 500
msec associated with 19% increase in ACE
(no further risk with QTc >500 msec)
Probst V et al. Study type: Inclusion criteria: BrS 1 endpoint: BrS assoc with SCN5A Poor genotype phenotype
Circ CV Gen 2009 multicenter families with at least 5 correlation for BrS SCN5A
20031634 retrospective family members genotype Results: BrS ECG present in 47% of
carries mutation carriers
Size: 115 Mutation carriers had longer PR and QRS
Exclusion criteria: N/A intervals
SCN5A mutations are not directly causal of
Br pattern ECG

Crotti L et al. Study type: Inclusion criteria: BrS 1 endpoint: Genotype results Brugada S Brugada: no genotype/phenotype
ACC 2012 Multicenter correlation
22840528 retrospective Results: 20% putative pathogenic
Size: 129 mutations, (95% in SCN5A; 5% other genes)
Yield similar with type 1 Brugada ECG only
(23%) and those with symptoms (17%)
Prolonged PQ interval > 200 msec: 38%
positive vs 11% if PQ < 200 ms, (OR 8, 1.5-
16)
Risgaard B et al. Study type: Inclusion criteria: Genetic 1 endpoint: Identify prevalence of ~10% of variants associated with
Clin Genet 2013 Exome variants of Brugada mutations associated with BrS in general BrS are present in Exome, raising
23414114 Sequencing Syndrome searched for in exome doubt about monogenic role in
Project (ESP) exome data BrS prevalence ~ 1:2000 to 1:100,000 pathogenicity of BrS
analysis Recommend using Exome data to
Exclusion criteria: N/A Results: 10% of variants identified in ESP, a establish gene frequency in
Size: 6258 frequency of 1:23 population

Data Supplement 40. Nonrandomized Trials Related to Congenital LQTS (Section 7.9.1.1.)
Study Acronym; Study 1 Endpoint and Results
Summary/Conclusion
Author; Type/Design; Patient Population (P values; OR or RR;
Comment(s)
Year Published Study Size & 95% CI)

196

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Garson AJ Circ Study type: Inclusion criteria: Age 1 endpoint: ACA or SCD for LQTS children QTc at presentation >0.60 highest
1993 (350) Retrospective <21y, QTc >0.44, during Mean followup 5 y. risk group
8099317 multicenter unexplained syncope, no difference between
seizures, ACA triggered by Results: Rx 68% BB, 8% other meds, LCSD propranolol and atenolol
Size: 287 emotion or exercise, or 2%, ICD 1% consider prophylactic treatment in
family Hx LQTS. Med treatment effective for symptoms in asymptomatic patients with QTc
Mean age presentation 76%, and for VEA 60% >0.44
8.8 y Symptoms in first mo of life high risk group:
61% symptoms 16% died.
9% ACA was first Asymptomatic patients with normal QTc and
symptom positive family Hx may be low risk group (no
genotyping results)
Exclusion criteria: N/A Predictors highest risk: symptoms at
presentation, propranolol failure
Hobbs JB et al. Study type: Inclusion criteria: 1 endpoint: ACA or SCD in adolescents with Risk factors: syncope, QTc 530
JAMA 2006 (351) Retrospective Adolescents in LQTS LQTS msec, males age 1012 y
16968849 multicenter Registry alive at age 10 y,
followed until age 20 y Results: 81 patients w ACA, 45 SCD
Size: 2772 Significant risk factors: recent syncope in
Exclusion criteria: N/A prior 2 y, HR: 11.7; QTc 530 msec HR: 2.3;
males age 10-12 y, HR: 4; males = females
ages 1320 y
Beta blocker therapy by 64% in patients
with syncope in last 2 y
Goldenberg I Study type: Inclusion criteria: 1 endpoint: LQTS with normal QTc: risk for Genotype positive patients with
JACC 2011 (340) Multicenter Genotyped patients with ACE: ACA or SCD normal QTc =25% of genotype
21185501 international LQTS: 3386 patients positive patients.
registry, Normal QTc: 440 ms Results: Normal QTc =14% of total LQTS 4% ACA/SCD with normal QTc vs
retrospective Prolonged QTc >440 ms patients in study. 15% if prolonged QTc
Unaffected: negative Normal QTc risk ACA/SCD =4%, lower than
Size: 469 genotype those with prolonged QTc (15%) but higher
than genotype neg family members.
Exclusion criteria: N/A Increased risk: mutation characteristics;
LQT1 vs LQTS 2, HR: 9.88; p=0.03;
Duration of QTc and gender important only
in those with prolonged QTc.

197

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Priori SG NEJM Study type: Inclusion criteria: 1 endpoint: LQTS risk of ACE age <40 y and Genetic locus and QTc
2003 (352) Retrospective Genotyped patients: before rx: syncope, ACA, sudden independent risk factors
12736279 LQT1 60%, LQT2 32%, deathbefore QTc risk factor for LQT1 and LQT2,
Size: 647 LQT3 8%, mean followup not LQT3
28 y Results: Incidence ACE: LQT1 30%, LQT2
46%, LQT3 42%. 13% ACA or sudden
Exclusion criteria: N/A deathbefore age 40 y,
Events highest among LQT2

Wedekind H Eur J Study type: Inclusion criteria: 1 endpoint: Recurrent syncope, ACA or SCD Risk predictors: QTc >500 msec,
Ped 2009 (339) Retrospective Genotype positive after dx LQTS. Mean followup 5.94.7 y prior syncope or ACA
19101729 single center probands, age 16 y LQT2 highest rate SCD vs other
LQTS: 89% LQT1, 2,3 Results: 92% treated: Followup: Propranolol
Size: 83 Mean QTc 51074 ms 79%, atenolol 20%, metoprolol 12%,
61% symptoms: syncope bisoprolol 8%, pindolol 2%; mexiletine 4%
49%, ACA 33%, SCD 18% ICD 8%, pacer 5%.
78% with BB rx 31% recurrent symptoms: 14% ACA or SCD;
syncope 86%
Exclusion criteria: N/A Significant predictors: QTc >500 ms, p=0.02,
HR: 2.9; 95% CI: 1.27.3; prior syncope HR:
4.04; 95% CI: 1.115, ACA HR: 11.7; 95% CI:
3.143.4, p<0.001
Jons C et al. JACC Study type: Inclusion criteria: LQTS 1 endpoint: Risk of ACE in LQTS patients Recurrent syncope during BB
2010 (353) Retrospective patients, QTc 450 msec with syncope treatment assoc with increased risk
20170817 International with syncope as first Severe = ACA, approp ICD shock, SCD of recurrent events
LQTS Registry symptoms BB failure highest in children and
20% with ICD Results: Lowest risk in patients with single females
Size: 1059 52 patients LCSD syncope before rx; intermediate risk:
multiple syncope before rx, HR: 1.8
Exclusion criteria: N/A Higher risk: syncope after BB rx: HR:3.6
p<0.001. Does not state how many patients
died/aca.
Barsheshet Circ Study type: Inclusion criteria: LQT1 1 endpoint: Risk for ACA/SCD vs. mutation LQT1 patients with C-loop
2012 (354) Retrospective genotyped patients, location in LQT1 mutations are at high risk for
22456477 observational mutations KCNQ1, ages ACA/SCD, and derive pronounced
birth-40 Results: 105 events: 27 ACA, 78 SCD benefit from b-blocker rx

198

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 860 Exclusion criteria: N/A C-loop mutations highest risk (HR: 2.75; 95%
patients CI: 1.295.86, p=0.009)
B-bl treatment sig greater risk reduction in C
loop mutations (HR: 0.12; 95% CI: 0.020.73,
p=0.02) vs all other mutations (HR: 0.82; 95%
CI: 0.312.13, p=0.68)
C-loop mutations showed sig reduction in
channel activation in response to b-
adrenergic stimulation
Vincent GM Circ Study type: Inclusion criteria: 1 endpoint: ACE (syncope, CA, SCD) in LQT 1 Risk for CA in compliant patients
2009 (355) Retrospective Genotype + LQT1 patients treatment with BB <<< non-compliant (OR:0.03; 95%
19118258 observational treatment with BB for CI: 0.0030.22, p=0.001)
minimum 2 y (unless Results: 75% asymptomatic. Beta-bl meds approp treatment
Size: 216 CA/SCD), median ACE 25%. for asxy patients, and symptomatic
followup 10 y. Median 5.5% CA/SCD (12 patients) after rx: 11/12 patients who have not had CA
age 26 y (476 y); non-compliant or on QT prolonging med. before b-bl rx.
73% symptomatic; prior None of 26 patients with prior CA had SCD Risk of CA/SCD on beta bl not
CA in 12% (26 patients). on beta-bl, one had CA. assoc with baseline QTc nor prior
Mean QTc 49548 ms Risk for CE reduced to 0.06 CE/y (0.050.07) syx nor gender
LQT1 patients with prior CA had
Exclusion criteria: N/A very low risk CA/SCD on BB
Moss AJ Circ 2000 Study type: Inclusion criteria: LQTS 1 endpoint: Recurrent CE on b-bl in LQTS For LQT 1 and 2, BB reduce risk
(344) Retrospective registry, Rochester, Highly symptomatic patients prior
10673253 observational patients treatment w BB Results: B-Bl significantly reduce risk LQT 1 to treatment at high risk for
age <41 y, 80% syncope and 2; recurrent events.
Size: 869 or ACA prior to rx. LQT 3: no effect LQT 3 patients: BB did not reduce
Atenolol, metoprolol, For symptomatic patients, HR 5.8 for risk
nadolol, propranolol. recurrent CE: 32% ACE within 5 y.
139/869 genotyped: LQT Prior syncope: HR: 3.1.
1(69), LQT 2 (42), LQT 3 Prior ACA, HR: 12.9 for ACA or sudden death:
(28) 14% recurrent CA.
Exclusion criteria: age
>41 y start rx

199

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Abu-Zeitone JACC Study type: Inclusion criteria: 1 endpoint: First cardiac event: syncope, All BB reduce risk of events,
2014 (356) Retrospective Patients in LQTS registry, CA, sudden deathafter starting b-bl without difference
25257637 multicenter Rochester, NY treatment In LQT 2 nadolol appeared
with BB: atenolol (441), Results: LQT 1: risk reduction 57% any b-bl, superior (HR: 0.40)
Size: 1530 metoprolol (151), no differential efficacy. For patients with recurrent events
propranolol (679), LQT2: nadolol only med with sig risk on beta-bl, propranolol offered least
nadolol (259), age <40 y, reduction (HR: 0.4) protection (HR: 0.52)
no AICD
Exclusion criteria:
simultaneous use of 2
beta Blockers
Goldenberg I JCE Study type: Inclusion criteria: 1 endpoint: Age related, gender and B-blockers reduced risk in LQT1
2010 (357) Retrospective Genotyped LQT1 (971) genotype specific risk factors for ACE and 2:
20233272 observational and LQT2 (422) patients (syncope, approp shock, ACA, or SCD) o LQT1 atenolol > nadolol
Multi-center in International LQTS Results: ACE LQT1 39%, LQT2 46% o LQT2 nadolol > atenolol
registry. Ages Birth-40 y. Risk for ACE:
Size: 1393 Ages 014 y, LQT1 genotype vs LQT2 ACA/SCD rarely occurred as
ICD 129 patients (LQT1 (HR: 1.49; 95% CI: 1.141.93, p<0.003); presenting symptom in patients
50, 9%; LQT2 79, 19%) males vs females (HR: 1.31, p=0.04) treatment with b-bl
Ages 1540 y, LQT2 vs LQT1, (HR 1.67; QTc 500 msec increased risk HR:
LCSD 31 patients, LQT1 95% CI: 1.312.13, p<0.001); females vs. 2.22.3
3%, LQT2 4% males HR: 2.58; 95% CI: 1.903.49, Syncope during b-bl treatment
p<0.001) assoc with increased risk ACA/SCD
Exclusion criteria: N/A QTC500 msec at increased risk in both Recommend BB therapy routinely
age groups: 014 y, HR: 2.3 (p<0.0001); to all high-risk LQT1 and LQT2
age 1540 y, HR: 2.22 (p<0.001) patients without contraindications
Treatment in LQT1: atenolol decreased as first rx
risk HR: 0.23; 95% CI: 0.080.67, 1 AICD therapy recommended for
p=0.008) nadolol was not associated those with syncope during b-bl
with sig risk reduction (HR: 0.4; 95% CI: therapy
0.141.16, p=0.09)
Treatment in LQT2: nadolol reduced risk
(HR: 0.13; 95% CI: 0.030.62, p=0.01);
atenolol did not (HR: 0.69; 95% CI: 0.32
1.49, p=0.34)
ACA or SCD rarely occurred during
treatment with beta-bl

200

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Patients with syncope during b-bl
treatment had rel high rate subsequent
ADA/SCD (>1 event per 100 pt-y.

Sauer AJ JACC Study type: Inclusion criteria: 1 endpoint: ACE: syncope, ACA, SCD Highest risk: females, QTc >500
2007 (358) retrospective Genotype positive LQTS between ages 18-40 y in LQTS msec, syncope after age 18 y
17239714 adults 18 y old LQT2 higher risk
8% prior ACA Results: Risk predictors: ACA or SCD: female QTc 499 msec did not contribute
gender HR: 32.68; QTc 500 ms HR: 3.34; to higher risk lethal event
Size: 812 Exclusion criteria: N/A QTc 550 msec HR: 6.35; syncope after age
18y, HR: 5.10
LQT2 33% recurrent ACE. LQT1 highest prior
events 34%.
BB reduced risk ACA, SCD by 60%; highest
benefit in QTc 500 msec, LQT1 and LQT2.
Steinberg C J Study type: Inclusion criteria: 1 endpoint: syncope, SCD, ACA, Bisoprolol (selective b-1
Interv Card EP 2016 retrospective Genotype positive LQT1 documented polymorphic VT LQT1 or 2, on antagonist) well-tolerated, and
(359) cohort (62%) or LQT2 (38%) BB shortened QTc similar to nadolol
27394160 treated with bisoprolol Median followup 3 y for bisoprolol and not powered to assess difference
Size: 114 52%, (59 patients), nadolol; 6 y for atenolol (p=0.03) in BB
nadolol 14%, (16
patients) or atenolol 34%, Results: Symptoms: 29%: syncope 27%, ACA
(39 patients) 3.5%, documented VT; ICDs 7%. Dosing:
59% females bisoprolol 5 mg, nadolol 6580 mg, atenolol
55 mg
Exclusion criteria: N/A Nadolol patients highest proportion of
probands vs bisoprolol (p=0.007)
QTc shortening greater with bisoprolol and
nadolol, vs. atenolol; QTc reduction greater
in nadolol vs. atenolol, similar to bisoprolol

201

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Cumulative incidence ACE 0.5%/pt-y.
ACA in one pt on bisoprolol; syncope in 2
patients with atenolol; no events with
nadolol
NO difference events bisoprolol 0.4% vs
other b-blocker 0.6%
Nannenberg EA Study type: Inclusion criteria: 1 endpoint: Using FTMR method to achieve Identify age ranges of highest risk
Circ CV Genetics Retrospective Genotype positive 6 Standardized Mortality Ratio(SMR) for specified inherited arrhythmia
2012 (336) single center, inherited arrhythmia (observed to expected mortality by genotype syndromes
22373669 Netherlands syndromes analyzed with and age in inherited arrhythmias Asymptomatic patients over age
Family Tree Mortality ranges may not require rx
Size: Ratio (FTMR): Results: LQTS1: in first 10 y of life SMR 2.9
LQT1,2,3; Brugada (1.55.1)
Syndrome, SCN5A LQTS2: age 30-39 y, SMR 4.0 (1.110)
overlap syndrome (LQT3, LQTS3: age 15-19 y, SMR 5.8(1.216.9)
BrS, conduction disease); SCN5A overlap syndrome: 20-39 y, SMR 3.8
RYR2 CPVT. (2.55.7)
CPVT: age 20-39 y, SMR 3.0 (1.36.0)
Exclusion criteria: N/A BrS: 40-59 y, SMR 1.79 (1.22.4), especially
males
Villain E EHJ 2004 Study type: Inclusion criteria: LQTS 1 endpoint: ACA or SCD in LQTS patients BB highly effective in children,
(360) retrospective in pt <18 y treated with <18yr old during followup median 7.5 y particularly in LQT1
15321698 single center BB, dx 1984-2002; 86% Double mutations or LQT2,3
genotype pos. Results: BB: nadolol 50 mg/m2/d given bid; higher risk
Size: 122 26 patients dx in first mo Propranolol 3-5 mg/kg/d, acebutolol 10 no LQT1 patient died while
of life; for others, median mg/kg/d., atenolol 50 mg/d, bisoprolol 10 receiving BB
age 6y at dx mg/d.
54% symptomatic Monitored at least yearly with ecg, exercise
probands test and/or holter, goal peak HR <130-150
bpm.
Exclusion criteria: N/A Symptomatic patients w longer QTc.
3 neonates died; one pt died after
pacemaker implantation.
One pt died after meds discontinued.
4.5% recurrent syncope. Cumulative event-
free survival 94%

202

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Moltedo JM Ped Study type: Inclusion criteria: 1 endpoint: Death, recurrent symptoms in Atenolol in twice daily dosing
Cardiol 2011 (361) retrospective Pediatric patients with young LQT1 ps treatment with atenolol effective in pediatric patients in
20960185 LQTS treated with during followup 5.44.5 y reducing events
Size: 57 atenolol. Assessing adequacy of beta-
Genotyping not available Results: Mean age dx 9 6 y, 60% females. blockade by blunting peak HR
Mean QTc 521 54 msec recommended
Exclusion criteria: N/A Mean dose atenolol 1.50.5 mg/kg/d twice Recurrent syncope occurred in
daily; dose titrated to achieve peak HR <150 patients with QTc >500 msec
bpm on holter and exercise.
+ family Hx sudden death22%. ICDs 10%
Symptoms 42%: VT: 18%, syncope 10%, ACA
7%, AV block 4%. One death, non-compliant
with meds.
Recurrent symptoms: 8%, 4 patients:
received ICD. All patients with recurrences
had QTc > 500 msec
6% side effects (1 pt) or inadequate heart
rate controlchange b-blocker
Schwartz et Aim: To assess Inclusion criteria: 162 1 endpoint: Cardiac events and on survival LCSD is associated with a
al.2004 (362) the long-term LQTS patients who free of cardiac events significant reduction in the
15051644 efficacy of LCSD underwent LCSD between incidence of ACA and syncope in
in a group of 1970 and 2002 were Results: Their QT interval was very high-risk LQTS patients when
high-risk identified. Among them, prolonged (QTc, 54365 ms); 99% were compared with pre-LCSD events.
patients. 15 underwent left symptomatic; 48% had a CA; and 75% of However, LCSD is not entirely
stellectomy that we those treated with BB remained effective in preventing cardiac
Study type: regarded as inadequate symptomatic. The average follow-up periods events including SCD during long-
Multicenter denervation and between first CE and LCSD and post-LCSD term follow-up.
global registry therefore insufficient were 4.6 and 7.8 y, respectively. After LCSD, The study population included the
therapy. Accordingly, the 46% remained asymptomatic. Syncope vast majority of LQTS patients
Size: 147 analysis is on the 147 occurred in 31%, ACA in 16%, and sudden treated with LCSD worldwide.
patients patients who underwent death in 7%. The mean yearly number of CEs Among 51 genotyped patients,
LCSD per patient dropped by 91% (p<0.001). LCSD appeared more effective in
Among 74 patients with only syncope before LQT1 and LQT3 patients.
Exclusion criteria: N/A LCSD, all types of CEs decreased significantly
as in the entire group, and a post-LCSD QTc
<500 ms predicted very low risk. The
percentage of patients with >5 CEs declined

203

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
from 55% to 8% (p<0.001). In 5 patients with
preoperative implantable defibrillator and
multiple discharges, the post-LCSD count of
shocks decreased by 95% (p=0.02) from a
median number of 25 to 0 per patient.
Bos JM Circ Study type: Inclusion criteria: LQTS 1 endpoint: LCSD for LQTS: ACE: syncope, 23% recurrent ACE after LCSD
Arrhythm Elect Single center patients undergoing LCSD ACA, SCD, approp ICD shock for VF
2013 (363) retrospective 2005-2010, mean QTc F/U 3.61.3 y.
23728945 52874 msec;
Size: 52 33% 1 prevention. Results: 23% recurrent ACE (not specified).
Mean age 14.110 y. 15% no reduction in events.

Exclusion criteria: N/A No recurrence in patients with b-bl


intolerance as indication (vs. recurrent
events). (0/12 vs 17/40, p<0.001)
Ptosis: 8%, pneumothorax 6%
Schneider, HE Study type: Inclusion criteria: LQT 5, 1 endpoint: LCSD for LQT, CPVT: ACE Reduction in ICD discharges
Clin Res Cardiol Retrospective CPVT 5, with recurrent LOS 3-9 d; followup median 2.3 y (0.63.9 y) 10% ACA
2013 (364) single center syncope, VT, ICD shocks Minor comps frequent
22821214 or ACA on BB.
Size: 10 Mean age 14 y (3.942 y). Results: Decrease in arrhythmia burden,
2 ICD pre-surg; 6 ICD at ACE
LSCD. No ICD discharges for VT
ACA: 10%
Exclusion criteria: N/A Horner syndrome 70%, 20% pleural effusion

Collura CA Heart Study type: Inclusion criteria: LCSD 1 endpoint: LCSD for LQTS and CPVT: ACE LCSD reduced shocks in 72%
Rhythm 2009 (365) single center 2005-2008, video- followup mean 17 mo during short term followup
19467503 retrospective assisted. Mean age 18% ineffective
9.19.7 y, (2mo-42 y) Results: 2 prev: ICD shocks eliminated 72%;
Size: 20 LQTS 12 geno +, 4 geno 18% ineffective
LQT; CPVT 2 2 prev 11, mean QTc 549 msec; 1 9, mean
QTc 480 msec.
Exclusion criteria: N/A
Hofferberth SC Study type: Inclusion criteria: LCSD 1 endpoint: ACE after LCSD: LQTS, CPVT, VF LCSD recommended in patients
JTCS 2014(366) single center 2000-2011. LQTS 13 Median followup 28 mo, (4131 mo) with recurrent symptoms refractory
24268954 retrospective to meds

204

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
(median age 8 y), CPVT 9 Results: 73% marked reduction in 27% recurrent symptoms, non-
Size: 24 (age 17 y), VF 2 (age 23). arrhythmia burden; 55% arrhythmia free. responders
27% persistent symptoms
Exclusion criteria: N/A
Chattha IS Study type: Inclusion criteria: 1 endpoint: Genotypic specific changes in End of recovery QTc >445 msec,
Heart Rhythm 2010 Retrospective Exercise testing done on QTc with exercise usually at 4 min of recovery,
(367) single center 3 groups: distinguished 92% of LQTS from
20226272 LQT1, LQT2, and controls Results: Changes in QTc: controls
Size: 75 LQT1: longer corrected QTc at peak and early Start of recovery QTc >460 msec
Exclusion criteria: N/A recovery correctly identified 80% of LQT1 and
LQT2: QTc increased during recovery 92% of LQT2
Controls: normal QTc during recovery
Aziz PF CAE 2011 Study type: Inclusion criteria: LQT1, 1 endpoint: QTc changes during exercise in QTc >460 msec at 7min of
(368) Single center LQT2, and controls LQTS recovery predicted LQT1 or LQT2 vs
21956039 retrospective undergoing cycle Results: LQT1 and LQT2 with sig increase in controls with 96% sensitivity, 86%
ergometer exercise QTc during recovery. specificity, 91% PPV.
Size: 158 testing Recovery delta QTc- (7 min-1 min) > 30 msec
predicted LQT2
Exclusion criteria: N/A
Laksman ZW JCE Study type: Inclusion criteria: LQT1 1 endpoint: LQT1 patients undergoing LQT1 patients with c-loop
2013 (369) Single center patients undergoing exercise: assess QTc and response to BB mutations did not increase QTc with
23691991 retrospective exercise testing; 28% with Results: no difference in QTc response exercise
C-loop mutations based on mutation location in LQT1; BB reduced supine, standing and
Size: 123 however, BB did not reduce QTc in c-loop peak exercise QTc
Exclusion criteria: N/A mutation patients

Sy RW Heart Study type: Inclusion criteria: 27 1 endpoint: CPVT outcomes: recurrent SVT occurred frequently (AF) and
Rhythm 2011 (370) single center patients with CPVT syncope, death or appropr shocks caused ICD shocks
21315846 retrospective Median age 35 y Patients presenting <21 y
33% presented 65% female Results: followup 6.25.7y appeared to have increased risk
<21 y CA 33%, syncope 56%, 63% exercise induced, 83% adrenalin death during followup
asymptomatic 11% induced; polymorphic VT more common Two deaths despite medications
Size: 27 ICDs in 15 patients with than bidirectional. and ICD therapies
CA or recurrent syncope SVT in 26%, (AF in 3, focal LA tach in 1)
on b-blockers; caused ICD shocks

Exclusion criteria: N/A

205

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
2 deaths, both in patients with ICDs: one VF
triggered by inappropriate shocks; one
incessant VT not-responding to ICD
4 appropr shocks; 19% inappropriate shocks
5 y risk ACE on b-blockers 4.9% all CPVT,
5.8% for RYR2 carriers
Spazzolini C JACC Study type: Inclusion criteria: LQTS 1 endpoint: Outcome of LQTS patients with ACA in first year of life are at very
2009 (371) Retrospective patients with ECG during ACA during infancy high risk of subsequent ACA/SCD
19695463 International first year of life during next 10 y of life
LQTS Registry Results: 70 patients events <1y: 20 SCD, 16
Exclusion criteria: N/A ACA, 34 syncope. BB not effective in preventing
Size: 212 Risk of ACE: HR <100, QTc 500 msec SCD/ACA in patients with prior ACA
ACA in first year: HR: 23.4 for ACA/SCD in
first 10y.
BB reduced risk in patients with syncope but
not ACA/SCD
Zhang C, et al. JCE Study type: Inclusion criteria: LQTS 1 endpoint: Identify major ACE (syncope, ADHD meds-stimulant or non-
2015 (372) LQT registry patients 1979-2003, with ACA, SCD) in patients with LQTS treatment stimulants-associated with
26149510 retrospective followup to 2015, treated with ADHD meds; mean followup 7.9y increased risk majory ACE,
with Attention particularly in mlaes
Size: 548 deficit/hyperactivity Results: 62% cumulative probablility of ACE
disorder (ADHD) in ADHD group, vs 28% in non-ADHD group.
medications Time dependent use increased risk, HR: 3.07,
p=0.03; increased riks in males, HR: 6.8
Exclusion criteria: other
LQT; patients with ICDs
Choy et al. 1997 Study type: Inclusion criteria: healthy 1 endpoint: Effect on QTUc from KCl after Potentially arrhythmogenic QT
(373) Double-blind subjects (12) and CHF quinidine or placebo. abnormalities during quinidine
9337183 comparison of (mean EF 17%) with age- treatment and in CHF can be nearly
potassium matched controls without Results: normalized by modest elevation of
infusion after CHF KCl was IV, 0.5 mEq/kg (to maximum of 40 serum potassium
quinidine and meEq) over 60-70 min resulted in
placebo Exclusion criteria: N/A normalization of quinidine-induced and CHF-
sequentially in related QTU prolongation
12 healthy
subjects.

206

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Also, study on
QTU in patients
with CHF and
age-matched
controls who
receive IV KCl

Size: 12
healthy, 8 CHF
plus 8 age-
matched
controls
Kannankeril P Study type: Inclusion criteria: N/A 1 endpoint: N/A Associated factors for drug
Pharmacol Rev Review Results: N/A induced LQTS; bradycardia,
2010 (374) Exclusion criteria: N/A hypokalemia; hypomagnesemia by
21079043 Size: N/A Lists drugs associated with torsades de modulating L-type calcium channel
pointes function
Drugs prolonging QT: block rapid
Genetic background-polymorphisms- may component of delayed rectifier
contribute to risk potassium current, IKr

Data Supplement 41. Nonrandomized Trials Related to Catecholaminergic Polymorphic Ventricular Tachycardia (Section 7.9.1.2.)
Study Acronym; Study 1 Endpoint and Results
Summary/Conclusion
Author; Type/Design; Patient Population (P values; OR or RR;
Comment(s)
Year Published Study Size & 95% CI)
Hayashi M Circ Study type: Inclusion criteria: CPVT 50 1 endpoint: ACE in CPVT patients: Higher risk for lack of BB, Hx ACA
2009 (347) single center probands, 51 family syncope, ACA, approp ICD shocks, SCD Prior syncope not associated with
19398665 retrospective members, age at dx 1510 increased risk
y. Results: followup 7.9 y
Size: 101 Symptoms 60% (61 8 y total event rate 32% total, 27% with b-bl,
patients), all probands, 22% 58% without b-bl. 8 y event ACA/SCD 13% (8
family members patients)
93% symptomatic <21 y old

207

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
77% detection of mutations: Increased risk: Absence BB HR: 5.54; 95% CI:
RYR2 CASQ2 1.1716.15, p=0.003), Hx ACA HR: 13.01;
95% CI: 2.4868.21, p=0.002); younger age
Exclusion criteria: N/A at dx (HR: 0.54/decade; 95% CI: 0.330.89,
p=0.02)
32% with events on b-blockers did not take
meds on day of event.
Nadolol: ACE 19%
Roston TM Circ Study type: Inclusion criteria: age <19 y 1 endpoint: ACE during followup in CPVT CPVT 25% recurrent events on
Arrh EP 2015 (375) multicenter dx with CPVT Treatment failure: syncope, CA BBcompliant, non-compliant,
25713214 retrospective Symptomatic 78%; 211 inadequate dosing
cohort treatment with meds: Results: Median followup 3.5y (1.45.3 y) High complications with ICDs
B-blockers: 91% Deaths 3% (6 patients): 2 patients receiving
Size: 226 AICD: 54% b-blocker; one previously asymptomatic
Flecainide 24%, calcium B-blockers: 25% recurrent events; 2% deaths
channel blockers Flecainide: 38% persistent VA, 16% failure
LCSD 8% (non-complaince, suboptimal dose);
LCSD: 18 patients: 16% complications; 67%
Exclusion criteria: N/A asymptomatic after rx; 11% recurrent VT,
5% CA (1 pt)
ICD: electrical storm 18%; 46% approp
shocks, 22% inappropriate shocks;
complications 23%
Chattha IS Study type: Inclusion criteria: Exercise 1 endpoint: Genotypic specific changes in End of recovery QTc >445 msec,
Heart Rhythm Retrospective testing done on 3 groups: QTc with exercise usually at 4 min of recovery,
2010 (367) single center LQT1, LQT2, and controls distinguished 92% of LQTS from
20226272 Results: Changes in QTc: controls
Size: 75 Exclusion criteria: N/A LQT1: longer corrected QTc at peak and Start of recovery QTc >460 msec
early recovery correctly identified 80% of LQT1 and
LQT2: QTc increased during recovery 92% of LQT2
Controls: normal QTc during recovery
Wilde AA NEJM Study type: Inclusion criteria: CPVT 1 endpoint: CPVT patients and LCSD: ACE LCSD does not preclude ICD
2008(376) Single center patients, treatment BB, after ICD implantation implantation
18463378 observational multiple ICD shocks: LCSD LCSD Reduced symptoms and
performed Results: no symptoms after LCSD shocks
Size: 3 RYR2 mutations

208

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: N/A LCSD recommended in CPVT
patients with symptoms on b-bl
therapy
Li J ATS 2008 Study type: Inclusion criteria: 11 1 endpoint: LQTS treatment with LCSD: LCSD reduced syncopal episodes
(377) Single center patients LCSD for LQT 2002- outcomes by 82%;
19022016 retrospective 2007, BB not tolerated or Mortality: 9.1%
refractory; followup time Results: 7/11 no symptoms;
Size: 11 3726 mos. 2 recurrent syncope; 1 SCD
Exclusion criteria: N/A
Collura CA Heart Study type: Inclusion criteria: LCSD 1 endpoint: LCSD for LQTS and CPVT: ACE LCSD reduced shocks in 72%
Rhythm 2009 single center 2005-2008, video-assisted. followup mean 17 mos during short term followup
(365) retrospective Mean age 9.19.7 y, (2mo
19467503 42y) Results: 2 prev: ICD shocks eliminated 18% ineffective
Size: 20 LQTS 12 geno +, 4 geno 72%;
LQT; CPVT 2 18% ineffective
Exclusion criteria: N/A 2 prev 11, mean QTc 549 msec; 1 9, mean
QTc 480 msec.
Schneider HE Study type: Inclusion criteria: LQT 5, 1 endpoint: LCSD for LQT, CPVT: ACE Reduction in ICD discharges
Clin Res Cardiol Retrospective CPVT 5, with recurrent LOS 39 d; followup median 2.3y (0.63.9 y) 10% ACA
2013 (364) single center syncope, VT, ICD shocks or Minor comps frequent
22821214 ACA on BB.
Size: 10 Mean age 14 y (3.942 y). Results: Decrease in arrhythmia burden,
2 ICD pre-surg; 6 ICD at ACE
LSCD. No ICD discharges for VT
Exclusion criteria: N/A ACA: 10%
Horner syndrome 70%, 20% pleural effusion
Hofferberth SC Study type: Inclusion criteria: LCSD 1 endpoint: ACE after LCSD: LQTS, CPVT, VF LCSD recommended in patients
JTCS 2014 (366) single center 2000-2011. LQTS 13 Median followup 28mo, (4131 mo) with recurrent symptoms refractory
24268954 retrospective (median age 8 y), CPVT 9 to meds
(age 17 y), VF 2 (age 23 y). Results: 73% marked reduction in 27% recurrent symptoms, non-
Size: 24 Exclusion criteria: N/A arrhythmia burden; 55% arrhythmia free. responders
27% persistent symptoms
Van der Werf C Study type: Inclusion criteria: 1 endpoint: reduction of VA in CPVT with Flecainide suppresses VA in CPVT,
JACC 2011 (378) multicenter Flecainide treatment for flecainide during exercise testing. Median up to 76%
21616285 retrospective genotype positive CPVT followup 20mo
patients, 8 European
Size: 33 centers prior to 12/2009;
209

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Results: Median age 25 y (768y); 73%
Exclusion criteria: N/A females
29/33 underwent exercise testing
Median dose flecainide in responders 150
mg (100300mg).
76% partial or complete suppression VA
with exercise (p<0.001); no worsening of VA
Appropr ICD shock in 1 pt, low serum flec
level
Watanabe H Study type: Inclusion criteria: 1 endpoint: Flecainide efficacy for Flecainide suppressed VA on
Heart Rhythm Single center Genotype negative CPVT suppressing VA in CPVT during exercise exercise testing in 75% of patients
2013 (379) retrospective with VA, syncope or ACA testing
23286974 Results: Mean followup 48 mo
Size: 12 Exclusion criteria: N/A Reduced arrhythmias 8/12 patients,
prevented VA 7/12
2/12 ACA/SCD, non-compliance
Priori S circ Study type: Inclusion criteria: CPVT 1 endpoint: CPVT genotype RyR2 vs Genotype positive RyR2 did not
2002(342) multicenter probands (30) underwent outcome correlate with VA, SCD, or response
12093772 retrospective genotyping; and 118 family to BB
members screened Results: RyR2 identified in 47% of
Size: 148 probands, and 9 family members, 4 clinically
Exclusion criteria: N/A silent
71% of gene positive were de novo;
29% familial: of familial, 75% asymptomatic,
55% VA on exercise test; 44% no syx or VA
on exercise testing
RyR2: events at younger age, males
increased syncope
Genotype positivity did not correlate with
VA, SCD, beta-bl rx

210

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 42. Nonrandomized Trials Related to Brugada Syndrome (Secction7.9.1.3)
Study Acronym; 1 Endpoint and Results
Study Type/Design; Summary/Conclusion
Author; Patient Population (P values; OR or RR;
Study Size Comment(s)
Year Published & 95% CI)
Gehi AK, et al. JCE Study type: Meta- Inclusion: Publications 1 endpoint: Identify risk predictors of BrS ACE risk increased with
2006 (333) analysis: retrieved 1/1990-3/2005 on adverse natural history in patients with prior syncope or SCD, RR: 3.24
16836701 30 prospective prognosis of patients Brugada ECG Males, RR: 3.47
studies on Brugada with a Brugada ECG: Spontaneous type 1 ECG, RR:
ECG Prospective cohort Results: 4.65
studies, >10 subjects, Risk increased with prior hx syncope or
Size: 1545 primary data on syncope, ACA, spontaneous type 1 Br ECG, and male
SCD, ICD shocks; gender
followup >6 mo and
>90% followup NOT sig risk factors: Fam hx SCD
SCN5A mutation, or inducibility by PES:
Exclusions: non-English; (not a risk factor but heterogeneity of
presence of cardiac studies)
disease
Somani R, et al. HR Study type: Inclusion criteria: 1 endpoint: Provocation of Brugada ECG Procainamide infusion provoked
2014 (380) Multicenter CASPER study of with procainamide infusion 15 mg/kg, Brugada ECG changes in ~7% of
24657429 prospective probands and first maximum 1 gm CASPER population.
degree relatives of
Size: 174 Unexplained cardiac Results: Mean age 47 yrs
arrest, SCD <60 y, VT or Procainamide: increased HR, prolongation
VF undergoing of QT.
cardioversion or Brugada ECG provoked in 12/174 = 6.9%
defibrillation, syncope 10/12 pts with ECG changes had SCN5A
with polymorphic VT mutation.

Exclusion criteria:
decreased LVEF, HCM,
CHD, overt Brugada ECG
pattern, prolonged QTc
Mizusawa Y, et al. Study type: Inclusion criteria: 1 endpoint: compare effects of fever and 3 aymptomatic patients
HR 2016 (381) multicenter Brugada S pts with fever drugs on BrS ECG developed VF/SCA during
27033637 retrospective 88 asymptomatic (79%) Subgroup of asymptomatc pts, (N=52), followup; 1/3 with spontaneous
26% SCN5A mutation serial ECGs BrS ECG,
211

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 112 Mean age 46 y followup
76% males Results: fever shortened PR, drug Paper is hard to interpret
challenge prolonged PR and QRS
Exclusion criteria: N/A
Drug challenge in 36 pts: ajmaline 24,
pilsicainide 7, flecainide 5
FINGER Study type: Multi- Inclusion criteria: 1 endpoint: ACE outcomes in BrS Low event rate in asymptomatic
Probst V Circ 2010 center registry, 11 Brugada Syndrome patients 0.5%/y.
(343) centers in Europe ECG spont (45%) or with Results: PES performed in 62%: 41% Inducibility w PES or family Hx
20100972 drug challenge. positive, higher in symptomatic patients SCD or SCN5A mutation not
Size: 1029 Median 45 y (35-55). 46% vs 37%, p=0.02. predictors of ACE
Hx ACA 6%, syncope 30%, PES performed in 369 asymptomatic Predictors of ACE: symptoms,
asymptomatic 64% (654 patients: 37% positive (137/369); 85% ACA, syncope, presence of ICD,
patients). (117/137) inducible asyx patients had ICD spont type 1 ECG.
SCN5A positive 22%. implanted Among asymptomatic patients:
ICDs implanted: 433/1029 patients (42%): 37% positive PES; of these 85%
Exclusion criteria: N/A of 433: 54 ACA (12.5%), 208 syncope had ICD implanted.
(48%), 171 asymptomatic (39%). 118/171 ICD implantation in
asymptomatic patients with ICD (69%) asymptomatic patients was
implanted due to positive EPS. significant in multivariable
analysis as predictor of ACE:
ACE 51: approp ICD shocks 44, SCD 7. HR:10.1; 95% CI: 1.758.7,
Mean ACE rate 1.6%/y: 7.7% in patients w p=0.01).
Hx ACA;1.9% w prior syncope; 0.5% in No independent predictive
asymp patients value of PES (p=0.09), males
Predictors: symptoms (p<0.001): ACA (HR: (p=0.42, spont type 1 ECG
11; 95% CI: 4.824.3, p<0.001), syncope (p=0.38) age (p=0.97)
(HR: 3.4; 95% CI 1.67.4, p=0.002),
ICD implantation (HR: 3.9; 95% CI: 1.4
10.6, p=0.007).
spont type 1 ECG (HR: 1.8;95% CI: 1.03
3.33, p=0.04);
NOT predictive: gender, family Hx SCD,
+PES (p=0.48), presence SCN5A mutation
Hiraoka M JE 2013 Study type: Inclusion criteria: 1 endpoint: Brugada S ages 18-35 y at Brugada outcomes in young
(349) Prospective single Brugada S patients ages dx, outcomes of VF or SCD adults vs presenting symptoms:
23702150 center 1835 y Followup 4327 mos.

212

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Mean age 306 y Events: VF 11.2%/y, syncope
Size: 69 Results: Based on presenting symptoms: 3.3%/y, asymptomatic 0.7%/y
No genetic testing VF 42%, syncope 12%, asymptomatic 2.5%
Not predictive: gender, family Hx SCD,
abnl SAECG, spontaneous vs drug-induced
Exclusion criteria: N/A ECG, inducible VT/VF

All ages 460 patients symptoms at


presentation vs outcomes:
VF 8.4%/y, Syncope 1.7%/y, asymptomatic
0.3%/y

PRELUDE Study type: Inclusion criteria: Age 1 endpoint: Predictive accuracy of PES PES did not predict high risk
Priori SG et al. JACC Prospective registry >18 y, BrS type 1 ECG for sustained VT/VF or approp ICD shock in Predictors: spontaneous type
2012 (382) spont (56%, 171/308) or Brugada S BrS ecg AND symptoms; f-QRS,
22192666 Size: 308 drug-induced, without VERP <200 msec
prior ACA; Results: PES performed at enrollment; VERP <200 msec was predictive:
followup every 6 mo. this data would only be obtained
21% with prior syncope Mean age 4512 y. at EPS.
(65 patients: 16/65 {25%} Cardiac arrest 4.5% (14/308), 13/14 NOTE that + PES used in decision
>1 syncope). resuscitated with ICD, EMS 1. to implant ICDs: 13/137 patients
PES positive in 41% (126/308); of these: (9.5%) with ICDs were
SCN5A positive 20% of single stimulation 5.5%, double 44.5%, resuscitated with ICD.
tested patients. triples 50%.
ICDs implanted in 137 patients (78% of Note 1/14 patients with VF had
(f-QRS = 2 or more spikes inducible patients {98/126} and 21% of only spont type 1 ECG and no
within QRS leads V1-V3: non-inducible patients {39/182}. prior syncope, neg family hx, neg
present 8.1%) Annual event rate 1.5%: EPS, VERP >200 msec but + SCN5A
mutation and received ICD after
Exclusion criteria: N/A Multivariable predictors: spont type 1 ECG EPS.
and Hx of syncope (HR: 4.20; 95% CI: 1.38 Only 1 pt without ICD had ACA: pt
12.79, p=0.012), Ventricular ERP < 200 had spont type 1 ECG, VRP < 200
msec (HR: 3.91; 95% CI: 1.0312.79, msec, and fQRS.
p=0.045),
QRS fractionation (HR: 4.94 95% CI: 1.54
15.8, p=0.007).

213

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Positive PES not predictive (HR: 1.03; 95%
CI: 0.343.16, p = 0.96)
Casado-Arroyo R Study type: Single Inclusion criteria: 1 endpoint: Long term trends Brugada S Brugada s: changes over time
JACC 2016 (383) center Compare BrS early period EPS Decrease in ACA over time as
27491905 retrospective 2002 vs. 2003-2014 presentation
Early: 165 Results: Early group more severe PES predictive in early group
Size: 447 Latter: 282 phenotype but not latter
ICDs: 48% early, 44% ACA 12% early, 4.6% latter, p =.005
latter PES positive 34% early, 19% latter,
p<0.001
Exclusion criteria: N/A Spontaneous type 1 ECG: early 50%, latter
26%, p=0.0002
Recurrent VA: early 19%, latter 5%,
p=0.007

Belhassen B et al, Study type: Inclusion criteria: 1 endpoint: Brugada S outcomes treated Brugada S: Class IA meds:
CAE 2015 (384) retrospective single Brugada S patients with IA drugs No deaths on quinidine;
26354972 center undergoing PES and Mean followup 11371 mo 40% of ACA patients remained
treated with Class IA arrhythmia free off AAD (3
Size: 96 drugs Results: Prior ACA 10, syncope 27, 59 treatment with quinidine for
Mean age 3916 y asymptomatic many years then discontinued rx
88% males PES: VF induced in 69% (100% of prior 38% side effects
ACA, 74% of syncope, 61% of
Exclusion criteria: N/A asymptomatic), PES RVA and RVOT in
most, 3 extrastimuli.
PES positive in 77% males, 9% females; in
88% with spont ECG vs 59% without spont
ECG.
Tested (60 patients) w quinidine (54),
disopyramide (2), both (4).
Quinidine prevented re-induction of VF in
90%; disopyramide 50%
30 Patients with neg PES were not
treated: all remained asymptomatic.
ICD implanted in 20 patients after PES
(30% of inducible VF patients):
complications 55% of patients.

214

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
4 died of non-cardiac causes.
Recurrent syncope: vasovagal 10, non-
arrhythmic 2.
2/96 had recurrent arrhythmia: both with
prior ACA; both discontinued quinidine
and had VF storms.
Nademanee K et al. Study type: Inclusion criteria: 9 1 endpoint: mapping and ablation of BrS shows delayed
Circ 2011(385) Retrospective single Brugada patients, RVOT in Brugada repolarization over anterior RVOT
21403098 center symptomatic with epicardium.
recurrent VF Results: Anterior aspect of RVOT Ablation normalizes ECG and
Size: 9 epicardium with late fractionate egms reduces VT/VF
median 4 episodes/mon; Ablation successful in 78% (7/9) VF not
median age 38 y; all with inducible, normalization of Brugada ECG in
ICDs 89%
Followup 206 mo, no recurrent VT/VF in
Exclusion criteria: N/A all patients off meds (except one on
amiodarone)
Sunsaneewitaykul B Study type: Inclusion criteria: BrS 1 endpoint: Ablation of zone of late Ablation of late activation zone
et al. JCE 2012 (386) Retrospective single patients EP mapping and activation in RVOT in RVOT may suppress VF storm
22988965 center ablation. between 8/07- and reduce VF recurrence
12/08 Results: Patients with VF storm: ablation
Size: 10 VF storm (4) and no VF modified Brugada ECG in 75% (3/4) and
storm (6) suppressed VF in all 4 during followup of
Exclusion criteria: N/A 1230 mo. RBBB in patients
Zhang et al. HR 2016 Study type: Two Inclusion criteria: BrS 1 endpoint: Brugada mapping and Ablation epicardial RVOT results
(387) center patients, 9 spont, 2 ablation of RVOT epicardium in normalization of Brugada ECG
27453126 retrospective induced and reduces VT/VF
Results: Normalization of spont Brugada ICD needed despite ablation
Size: 11 Exclusion criteria: N/A ECG pattern in all
73% free of VT/VF at 2511 mo
Brugada J et al. Circ Study type: Inclusion criteria: BrS, 1 endpoint: Epicardial mapping and Ablation may eliminate
A E 2015 (388) Single center spont ECG, median age ablation RVOT in Brugada spontaneous Brugada ECG pattern
26291334 retrospective 39 y Results: Ablation resolved spontaneous
Size: 14 Exclusion criteria: N/A Brugada ECG
5 mo, no recurrence

215

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
McNamara DA Study type: nclusion criteria: 1 endpoint: All-cause mortality, ACE in Decreased mortality in patients
Cochrane Database Cochrane search for patients >18 y, ion BrS and ICD randomized to ICD in BrS: 9-fold
Syst Rev 2015 (389) randomized trials of channelopathies, reduction
ICD vs medical randomized to ICD vs Results: 2 studies identified, Brugada
treatment ion medical rx, identified 2 Syndrome, same authors. Brugada patients with prior
channelopathy studies including Brugada ICD: assoc with decreased risk mortality ACA: ICD treatment reduced
patients RR: 0.11; 95% CI: 0.010.83) mortality
Size: 86 Adverse events higher in ICD: 28% vs 10%,
Exclusion criteria: N/A RR: 2.44; 95% CI: 0.926.44)
Non-fatal ACE higher in ICD: 26% vs 0%,
RR: 11.4; 95% CI: 1.5783.3)
Delise P et al. EHJ Study type: Multi- Inclusion criteria: Type 1 1 endpoint: predictors in Brugada S of Combining 2 risk factors was
2011 (348) center prospective Brugada ECG: ACE (approp ICD shocks, sudden death) useful risk stratification:
20978016 spontaneous 54%, drug- Spontaneous type 1 ECG
Size: 320 induced 46%. Results: Median followup 40 mos (IQR Family Hx sudden death, syncope,
2067) positive PES
Median age 43 y. 5.3 % MACE (17 patients): VF on ICD (14), MACE occurred only in patients
Males 81% sudden death3 with 2 risk factors
MACE occurred in 10.4% of symptomatic MACE event rates:
Asymptomatic 66%, and 2.8% of asymptomatic patients 3.0%/pt/yr in symptomatic,
syncope 33% (p=0.004) 0.8%/pt/yr in asymptomatic
ICDs implanted in 34%(110 patients) PES can be useful in patients
No prior ACA PES performed in 245 (76%): positive in with spontaneous type 1 ECG and
50% of symptomatic and 32% of no other risk factors; may be
asymptomatic patients. helpful to identify low risk
Exclusion criteria: N/A MACE in 14% of positive PES, 0% of patients
negative, 5.3% of no EPS: positive
predictive values 14%, negative pred value
100%
VF occurred in 15.5% of patients with
inducible VF using doubles, 8.6% of triples
Combination of risk factors most
significant: spont ECG, family Hx sudden
death, syncope, positive EPS: no events
occurred in patients without any of above
or with only one risk factor.

216

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Spontaneous type 1 ECG: if additional risk
factors, 30% MACE (p<0.001)
Sieira J et al. Circ Study type: Single Inclusion criteria: 1 endpoint: Event-free survival in Brugada S: Positive PES
Arrhyth EP 2015 (390) center Asymptomatic patients Brugada S. predictor of adverse events, HR:
26215662 retrospective type 1 BrS ECG, spont Mean followup 7359 mo. 9.1.
(11%) or drug-induced. Event free survival 95.4% at 10
Size: 363 Mean age 40.917 y, Results: PES positive in 10% (32 patients) and 15 y
55% males. ICDs implanted 17% (61 patients), 6
321 patients underwent approp rx.
PES. Event free survival: 99% 1 y, 96% at 5 y,
22% genotype + SCN5A. 95.4% at 10 and 15 y.
Arrhythmic events: 9, annual incidence
Exclusion criteria: N/A 0.5%
Multivariate analysis: Positive PES only
significant predictor (HR: 9.1, 95% CI: 1.8
46.8, p<0.01)
Konigstein M et al. Study type: Inclusion criteria: 1 endpoint: Outcomes of non-cardiac Non-cardiac drug induced type
Heart Rhythm 2016 multicenter Brugada database non- drug-induced BrS 1 Brugada ECG:
(391) retrospective cardiac drug-induced 26% VF/pulseless VT
27131070 Brugada patients; each Results: By definition: spontaneous type 13.5% mortality
Size: 74 with 5 healthy controls 1 ECG:
Mean age 3916 y. 49% psychotropic meds (lithium,
77% males amitriptyline), 27% anesthetic/analgesic,
24% other; of total, 20% propofol
Exclusion criteria: N/A occurred predominantly in adult males,
frequently due to drug toxicity, occurs late
after onset of treatment
Off-drug ECGs: 33% type IIC Brugada ECG
Sroubek J et al. Circ Study type: Inclusion criteria: BrS 1 endpoint: CA or appropriate ICD shock Positive PES associated with
2016 (392) Systematic review patients without ACA in Brugada S. increased risk ACE during
26797467 and pooled analysis who underwent PES followup; induction with 12
of prospective Mean age 44.9 13.3 yrs; Results: PES induced sust VEA (40%).with extrastimuli associated with
observational 79% male; 53% spont up to triple extrastimuli in 527 patients higher risk.
studies type 1 ECG (2%, single; double 18%; triples 28% Specificity of induction as risk
AICDs implanted in 576 patients: 77% of predictor decreased with triple
Size: 8 studies, Prior Syncope 33%; ICD implanted in PES positive patients VEST
1312 patients

217

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: N/A 65 patients experienced ACE during Negative PES did not identify
median followup 38 mo: 5 CA, appropriate low risk individuals
ICD shock 60. Annual event rates varied based
Positive PES assoc with increased risk ACE: on syncope, spontaneous type 1
HR: 2.66, 95% CI: 1.444.92, p <0.001); ECG, and positive PES:
greatest risk in those induced with single Asymptomatic patients with
(HR: 1.99, 95% CI: 0.527.68, p=0.32); or spont type ECG and positive PES:
double extrastimuli (HR: 2.55, 95% CI: annual incidence 1.70 (0.733.35)
1.344.88, p=0.005), vs. triples (HR: 2.08, Aymptomatic patients with drug
95% CI: 0.984.39, p=0.06) ind ECG and + PES: annual
Clinical variables useful: annual event incidence 0.45 (0.012.49)
rates for no syncope, drug induced type 1 Clinical factors important
ECG: 0.27% (95% CI: 0.070.68); Positive determinants of risk: syncope;
syncope and spont type 1 ECG 3.22%; spont type 1 ECG
(95% CI: 2.234.5) Asymptomatic patients with
Highest risk: + syncope, spont type 1 ECG: drug induced ECG patterns: PES
neg PES HR: 2.55; 95% CI: 1.583.89; may not be warranted
positive PES HR: 5.6; 95% CI: 2.989.58 Symptomatic patients:
Annual incidence rates of CA or VT: increased risk with positive PES,
Asymptomatic, spont type 1 ECG: annual but risk exists with neg PES:
events 1.04 (95% CI: 0.611.67): positive higher if spont type 1 ECG: ? value
PES 1.70 (95% CI: 0.733.35); negative PES of PES
0.78 (95% CI: 0.361.47)
Asymptomatic, drug ind ECG: overall 0.27,
neg PES 0.23 (95% CI: 0.050.68), pos PES
0.45 (95% CI: 0.012.49)
Spont type 1 ECG: asymptomatic, with
neg PES: annual event incidence 0.78%
(95% CI: 0.361.47); pos PES 1.70 (95% CI:
0.733.35).
Prior syncope and neg PES 2.55% (95% CI:
1.583.89); Positive PES 5.60 (95% CI:
2.989.58)
Drug induced ECG: asymptomatic: neg
PES 0.23% (95% CI: 0.050.68); positive
PES 0.45 (95% CI: 0.012.49); prior
syncope and negative PES 1.29 (95% CI:

218

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
0.522.67); positive PES 1.96 (95% CI:
0.405.73)
Sieira J et al. Heart Study type: Single Inclusion criteria: 1 endpoint: Brugada outcomes in BrS Females:
2016 (393) center Women with BrS, women, mean followup 73 mo Less severe than males, less
26740482 retrospective spontaneous 8%, or spont type 1 ECG
induced Results: Mean age 41.5 17.3 y Event rate 0.7%/y (males
Size: 228 women = 42% of Brugada population 1.9%/y)
Exclusion criteria: N/A Spontaneous type 1 ECG 7.9% vs males Higher risk: prior ACA, SND
23%, p<0.01
ICD implanted in 28%, event rate 0.7%/y
vs 1.9% males
Priori S et al. Circ Study type: Inclusion criteria: 1 endpoint: Brugada risk stratification for Multivariable risk predictor:
2002 (394) Multicenter Brugada S with ECG SCD spontaneous ST elevation V1-V3
11901046 retrospective changes, spont (51%) or PES performed in 86 and Hx of syncope
induced Syncope without spontaneous
Size: 200 Results: SCN5A identified in 22% ST elevation not a risk factor
130 probands probands, 46% of family members PES not predictive
Exclusion criteria: N/A Risk analysis: gender; ECG, family hx, Mutation carriers without
mutation status, symptoms phenotype: low risk
Syncope without ST elevation on baseline
ECG: not a risk
Syncope AND ST elevation: increased risk
SCD, HR: 6.4, p<0.002
Fauchier L et al. IJC Study type: meta- Inclusion criteria: 1 endpoint: utility of PES in Brugada S: Inducibility of VT in Brugada S
2013 (395) analysis Brugada S patients adverse event = sust VT/VF, appropriate patients with syncope or
23642819 undergoing PES ICD shock, sudden death) asymptomatic may identify an
Size: 1789 ACA 11%, syncope 31%, increased risk of subsequent
asymptomatic 57% Results: Inducible VT/VF associated with events
higher risk arrhythmic event in patients
Exclusion criteria: N/A with prior syncope (OR: 3.30, 95% CI:
1.686.51, p=0.0006) and in asymptomatic
patients (OR: 4.62, 95% CI: 2.149.97,
p<0.0001)
Rodriguez-Manero Study type: Inclusion criteria: BrS 1 endpoint: ICD usage and comps in BrS:
M et al. Heart Rhythm retrospective multi patients with implantable Brugada S. ICD approp use in ~14%
2016 (396) center ICD followup mean 69 54 mo Monomorphic VT in 4.2%
26538325 1993-2014 Results: 13.7% at least one approp rx

219

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 834 mean age 4513.9 y Monomorphic VT recorded in 4.2% (35 Successful ablation in 80% of 10
24% women patients), sensitive to anti-tach pacing in patients with outflow tract VT
43%
Exclusion criteria: N/A Monomorphic VT from RVOT 6, LVOT 2,
BBR 2 successfully ablated in 80%
Sacher F et al. Circ Study type: Inclusion criteria: BrS 1 endpoint: ICD outcomes in BrS, Approp ICD shocks more
2013 (397) Retrospective patients with ICD followup mean 7742 mo prevalent in symptomatic BrS;
23995538 multi-center Mean age 4613 y Asymptomatic patients had
ACA 31, syncope 181, Results: appropriate shocks 12%, approp shocks 1%/y
Size: 378 asymptomatic 166 Shock rates highest for ACA patients Optimal programming may
(48%), syncope 19%, 12% asymptomatic reduce inapprop shocks
Exclusion criteria: N/A Inaapropriate shocks 24%; due to lead Lead failure a significant
failure, SVT, T wave oversensing or sinus problem
tach. Lead failure 29%
Rosso R et al. Isr Study type: Inclusion criteria: BrS 1 endpoint: Followup efficacy and comps Appropriate shocks occurred
Med Assoc J 2008 retrospective multi- patients with ICD of ICD in Brugada; only in symptomatic patients with
(398) center, 12 centers, Mean age 44.1 y followup 4535 mo prior ACA
18669142 1994-2007 VF inducibility did not predict
Exclusion criteria: N/A Results: Symptoms 71%: ACA 19%, approp shocks
Size: 59 syncope 53%, inducible VF in High complication rate
asymptomatic patients 24%, family Hx SCD
0.5%.
Appropriate shocks 8.4%, all with prior
ACA
Comps 32%
Inappropriate shocks 27%
Psych problems 13.5%, mainly related to
inappropriate shocks
Conte G et al. JACC Study type: Inclusion criteria: BrS 1 endpoint: Long term followup ICD in ACA and VT inducibility on EPS
2015 (399) Prospective single patients with ICDs BrS, mean followup 8457 mo were multi-variate predictors of
25744005 center appropriate shocks
Exclusion criteria: N/A Results: Spontaneous VA in 17%. Appropriate shocks occurred in
Size: 176 Appropriate shocks 15.9% 13% of asymptomatic patients
Inappropriate shocks 18.7%
Electrical storm 2.3%
SCN5A mutation (22%) did not correlate
with approp shocks

220

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Miyazaki S et al. AJC Study type: single Inclusion criteria: 1 endpoint: Brugada S ICD outcomes Brugada S + ICDs:
2013 (400) center Brugada S patients with Median followup 76 mo Complications 37%
23433764 retrospective ICD
Mean age 4812 y Results: Complications 37%: device related
Size: 41 93% males 20%, inappropriate shocks in 24%
Appropriate shocks: 12%
Exclusion criteria: N/A
Takaqi M et al. Study type: Inclusion criteria: 1 endpoint: ACE documented VT or SCD ICD implantation in Brugada:
Heart Rhythm retrospective single Brugada S patients in Brugada S with ICD Higher events in IIa vs IIb
2014(401) center undergoing ICD Mean followup 6031 mo Spontaneous type 1 ECG AND
24981871 implantation, syncope useful for identifying
Size: 213 Mean age 5314 y Results: indications classified as intermediate risk
Males 93% IIa (66): spontaneous type 1 ECG and Hx
of cardiac syncope, or
Exclusion criteria: N/A IIb (147): spont or drug induced type ECG
and inducible VF by PES.
Event rates: IIa 12%, 2.2%/y;
IIb 3%, 0.5%/y p=0.01

Data Supplement 43. Nonrandomized Trials Related to Early Repolarization J-wave Syndrome (Secction 7.9.1.4)
Study Acronym; Study 1 Endpoint and Results
Summary/Conclusion
Author; Type/Design; Patient Population (P values; OR or RR;
Comment(s)
Year Published Study Size & 95% CI)
Rosso R et al. JACC Study type: Inclusion criteria: 1 endpoint: Assess frequency of ER on J point elevation occurs more
2008 (398) Retrospective Idiopathic VF patients ECG vs controls frequently in idiopathic VF
18926326 single center compared with 123 patients than healthy controls
age/gender matched Results: ER more common among VF Athletes intermediate frequency
Size: 45 controls. patients, 42% vs 13%, p=0.001 of J point elevation between
Mean age 3815 y, 71% J point elev in inferior leads: 27% vs 8%, normal adults and idiopathic VF
male p=0.006 patients
2/45 dx with Brugada J point elev in leads I-aVL 13% vs 1%, ST segment elevation or QRS
p=0.009 slurring did not add diagnostic
Exclusion criteria: N/A J point elev in V4-V6 equal among values
groups, 6.7 vs 7.3%

221

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Males more often had J point elev vs
females; young athletes more frequent
than controls but less than VF patients

Haissaguerre M, et Study type: Inclusion criteria: 1 endpoint: Recurrent VF >3 episodes Recurrent VF high: 40% with
al. JACC 2009 (402) multicenter cohort Idiopathic VF survivors mult episodes in 27%
19215837 with ER assessed for Results: overall 27% with multiple (>3 Meds not effective other than
Size: 122 recurrent VF episodes) of recurrent VF quinidine or hydroquinindine (9
Inducible VF 28% in entire cohort pts)
All pts had AICDs Pts with >3 episodes recurrent VF:
implanted inducible VF 48%, p<0.01, prior syncope
58%, p<0.001 compared with pts with <3
Mean age of diagnosis 39 episodes of recurrent VF. Anti-
y arrhythmic meds not highly effective in
preventing recurrent VF
Exclusion criteria: 1 death due to refractory VF
Tikkanen JT ET AL. Study type: Inclusion criteria: ECGs 1 endpoint: Death from cardiac causes; ER pattern in inferior leads of
NEJM 2009 (403) retrospective obtained in general 2: death from any cause and from ECG is associated with an
19917913 community based population reviewed, arrhythmia before end of 2007; mean increased risk of death from
screen of ECGs in followup 3011 y. cardiac causes in middle-aged
Finnish population Exclusion criteria: N/A adults
1962-1972 Results: Prevalence J point elev of at ER transmural heterogeneity in
least 0.1 mV: 5.8%: inferior leads 3.5 %, vent repolarization, increases risk
Size: 10864 70% male; Lateral leads 2.4%, 58% male during cardiac ischemia
J point elev at least 0.2 mV inferior leads
0.3%, lateral 0.3%
Cardiac death: ER patients (RR: 1.28, 95%
CI: 1.041.59, p=0.03); arrhythmia death
J point elev 0.2 mV: cardiac death RR:
2.98, 95% CI: 1.854.92, p=0.01;
arrhythmic death RR: 2.92, 95% CI: 1.45
5.89, p=0.01
QTc (RR: 1.2, 95% CI: 1.021.42, p=0.03)
and LVH (RR: 1.16, 95% CI: 1.051.27,
p=0.004) weaker predictors cardiac
death

222

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Sinner MF et al. Study type: 3 Inclusion criteria: 452 1 endpoint: Combined meta-analysis Unable to reliably identify
Heart Rhythm 2012 community based patients with ER failed to reach genome wide significance genetic variants predisposing to ER
(404) ECG cohorts underwent genome wide
22683750 Size: 7482 association studies Results: ER: 70% male
Exclusion criteria: N/A
Adhikarla C et al. AJC Study type: Inclusion criteria: ER > 1 endpoint: assess changes in ER on ER pattern lost in over half of
2011 (405) retrospective 0.1 mV with ST segment ECG during 10 y followup young male cohort over 10 y
21907947 Screening ECGs on elevation, J wave as period, not related to death
veterans for ER upward defection, slurs as Results: 122/244 patients had second
1987-99 delay on R wave ECG
downstroke: first 250 ER persisted in 38%; most no longer filled
Size: 29281 patients selected. Mean criteria.
4210 y

Exclusion criteria: other


ECG abnormalities
Siebermair J, et al. Study type: Single Inclusion criteria: 1 endpoint: Appropriate VF shocks on Recurrent VF high: 43%
Europace 2016 (406) center Idiopathic VF survivors ICD in idiopathic VF pts; compare ER to Recurrent VF higher in ER
26759124 retrospective assessed for ER and ICD non-ER patients
interventions during High incidence AF in VF survivors
Size: 35 follow-up median 8.8 y Results: overall 43% recurrent VF after
median 6.6 yrs.
Exclusion criteria: N/A VF more frequent in ER patients: (HR:
3.9, 95% CI: 1.411.0, p=0.01)
40% inappropriate shocks: 66% due to AF
Cheng YJ, et al. JAHA Study type: meta- Inclusion criteria: studies 1 endpoint: risk of SCA, cardiac death, Early repolarization associated
2016 analysis assessing link between ER death any cause associated with early with absolute risk increase of
27671315 and risk of SCA, cardiac repolarization pattern on ECG 139.6 additional SCAs/100,000 pt
Size: 16 studies death, and eath from any y and responsible for 7.3% of SCA
including 334,524 cause Results: Increased risk of SCA (RR:2.18, in general poulation
patients identified 95% CI: 1.293.68), and cardiac death
Exclusion criteria: N/A (RR: 1.48, 95% CI: 1.062.07) in patients
with early repolarization.
Increased risk predominantly in Asians
and whites but not African Americans.
J-point elevation in inferior leads,
notching configuration, and harizaontal

223

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
or descending ST segement connote
higher risk.
Tikkanen JT et al. Study type: Inclusion criteria: 1 endpoint: Prevalence of ER in SCD vs Higher prevalence of ER in SCD
Circ AE 2012 (407) Retrospective Prevalence of ER in survivors of acute ischemia ischemic patients than in survivors
22730409 population based Baseline ECGs of 432 of acute coronary event
consecutive cases of SCD Results: Prevalence ER 0.1 mV in at ER increases vulnerability to
Size: 432 due to ischemia least 2 inf or lateral leads: 14.4% cases vs fatal arrhythmia during acute
compared with 532 7.9% controls. myocardial ischemia
survivors of acute ER with horizontal or descending ST
ischemic event segment assoc with SCD 10.2% vs 5.3%,
p=0.004; ER with ascending ST NS.
Exclusion criteria: N/A SCD patients younger, more often male,
smokers, lower BMI, elevated HR,
prolonged QRS complex, lower
prevalence of Hx of CVD

Junttila MJ et al. Study type: Inclusion criteria: 1 endpoint: Sustained VT or VF, Inferior ER without other QRS
Heart Rhythm 2014 Community based arrhythmic outcomes and arrhythmic death, non-arrhythmic cardia morphology changes predicted
(408) ECGs Finnish cardiac deaths in patients death, AF, CHF, CAD; mean followup occurrence of VT-VF but not non-
24858812 population, mean with ER on community 3011 y arrhythmic cardiac events
448 yrs screening Suggests ER sign of increased
Results: Inferior ER 3.5% prevalence: vulnerability to ventricular
Size: 10,846 Exclusion criteria: N/A predicted VF-VT events (N=108), HR: 2.2 tachyarrhythmias
(1.14.5, p=0.03), not not nonarrhythmic
cardiac death, CHF, or CAD
Inferior ER predicted arrhythmic death in
cases without other QRS abnormalities
(HR: 1.68, 95% CI: 1.12.58, p=0.02) but
not in those with coexisting
abnormalities in QRS morphology (HR:
1.3, 95% CI: 0.861.96, p=0.22)

224

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 44. Nonrandomized Trials Related to Short-QT Syndrome (Secction 7.9.1.5)
Study Acronym; 1 Endpoint and Results
Study Type/Design; Summary/Conclusion
Author; Patient Population (P values; OR or RR;
Study Size Comment(s)
Year Published & 95% CI)
Gaita F et al. JACC Study type: single Inclusion criteria: 1 endpoint: Prolongation of QTc with Hydroquinidine prolonged QTc
2004 (409) center retrospective Symptomatic patients with medications and resulted in non-inducible VF
15093889 QTc <380 undergoing drug use dependent block fast inward
Size: 6 testing. One prior ACA age Results: Flecainid, sotalol, ibutilide, Na, blocks rapid IKr and IKs, IKATP,
6 y. hydroquinidine tested. Ito.
PES 5 adult patients: 4/5 Only hydroquinidine prlonged QTc from
inducible VF. 26312 to 36325, prolonged VERP to
5 adults received ICDs. 200 msec, and no VF induced.

Exclusion criteria: N/A


Giustetto C et al. Study type: Inclusion criteria: Short 1 endpoint: outcomes with AICD or Short QTS may be a cause of SCD
EHJ 2006 (51) Retrospective single QTc 340 msec and hydroquinidine in infancy
16926178 center personal or family Hx of
CA. 73% males. Results: Median age dx 30 yrs (4-80); Hydroquinidine may be
Size: 29 62% symptomatic: syncope 24%, AF 31%. proposed in children or patients
Exclusion criteria: N/A 34% ACA (10 patients); 2/10 had CA in not suitable for AICD
infancy. In 28% ACA was initial symptom.
AICD implanted in 14; 10 hydroquinidine. PES sensitivity 50%
Median followup 23 mo (9-49), one pt
with appropriate ICD shock. No pt on
hydroquinidine had SCD or syncope.

PES 18/29: VERP 140-180 msec. VF


induced in 61% (11/18); 3/6 with
documented VF had inducible VF:
sensitivity 50%. AERP CL 600: 120-180
ms, mean 157.
Gollob MH et al. Study type: Inclusion criteria: review 1 endpoint: review reported cases of Gollob criteria for SQTS, 4
JACC 2011 (410) Medline database details of reported cases of Short QTS: 61 cases worldwide points very likely
21310316 search SQTS QTc duration <370, <350, <330
Results: Increased in males: 75% mean J point-Tpeak <120 msec
Size: 61 Exclusion criteria: non- QTc 397 msec, 248381 msec in Clinical hx: ACA, SCD, AF,
English journals symptomatic cases. unexplained syncope;

225

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Family hx; Genotype results
Giustetto C et al. Study type: Inclusion criteria: 1 endpoint: syncope, CA or approp ICD SQTS assoc with SCD in all ages
JACC 2011 (53) retrospective multi- European Short QT shocks SQTS Symptomatic patients have high
21798421 center Registry patients with QTc risk of recurrent arrhythmic events
360 msec with Hx sudden Results: Mean Followup 6427 mo. Patients treated with
Size: 53 death, ACA, syncope; Median age 26 y (IQR 1739). 62% Hydroquinidine did not have
patients with QTc 340 symptomatic: 32% with ACA (13 patients) arrhythmic events
msec included without or sudden death(4), syncope 8, AF 6, Asymptomatic patients: no
symptoms. palps 13. CA/ICD shocks.
75% males. Age at CA 3 mos62 y. PES not sensitive
Family Hx SCD/CA (11). Males: >90% of CA occurred between
Genotype positive 23% of 14-40 yrs.
probands: HERG in 4 Prevalence CA males 35%, females 30%.
families (N588K in 2, AICD in 24, hydroquinidine in 12.
T6181 in 2; CACNB2b in 11/12 with prior CA received ICD: 2
one family) approp ICD shocks. 58% complications of
. ICD, inapprop shocks due to T wave
Exclusion criteria: N/A oversensing 4/14.
PES: 28 patients. VERP CL 600-500: mean
166 msec. AERP 166 msec. VF induced in
16/28: 3/28 with prior CA = sensitivity
37%, NPVs 58%.
Overall event rate 3.3%/y: 4.9% in
patients without AA drugs.
Asymptomatic patients: 27. ICD
implanted in 9 due to + family Hx or
induced VF. Two long term quinidine.
One syncope; 2 nonsust VT on ICD.
Villafane J et al. Study type: Inclusion criteria: patients 1 endpoint: ACE in short QT; Assess modified Gollob score >5
JACC 2013 (411) Multicenter <21 y old with short QTc Gollob score associated with likely clinical
23375927 retrospective <360 msec. Mean followup 6 y. events
Median age 15 y Results: Symptoms 56%: ACA 24%, High rate inappropriate shocks
Size: 25 syncope 16%
Exclusion criteria: N/A 84% personal or family Hx ACA/SCD
24% genotype +
AICD 11: 2 approp shocks; 64%
inappropriate shocks

226

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
10 patients med rx: quinidine
Gollob score <5 remained event free
(excluding patients for symptoms)
Mazzanti A et al. Study type: Inclusion criteria: Short 1 endpoint: SQTS patients followed for SQTS highly lethal at young age
JACC 2014 (412) Registry QTS: asymptomatic 340 median 56 mo 11% genotype positive
24291113 msec, or QTc 340360 Prior ACA predicts recurrent CA:
Size: 73 msec Plus ACA, family Hx Results: 84% male Mean age 2615 y, recommend ICD for these patients
SCD or family Hx SQTS QTc 32922 msec. 40% presented with Gollob score did not predict risk
53% symptomatic at ACA, range 1 mo41 y.
referral CA during sleep 83%, 17%
Exclusion criteria: N/A emotion/exertion
Rate CA 4% first yr of life, 1.3%/y
between 20-40 y.
Probability first occurrence CA by 40 y:
41%.
ACA only predictor of recurrence:
p<0.0000001
Iribarren C et al. Study type: Inclusion criteria: 1 endpoint: Prevalence, risk of death QTc 300 msec: 2.6 fold
Ann Noninv ECG Retrospective Screened 6,387,070 ECGs associated with Short QT during 8.3 y increased risk death
2014 (413) in population of 1.7 million median followup
24829126 Size: 1026 persons for QTc 300 msec
Results: Prevalence 2.7/100,000, or
Exclusion criteria: N/A 1/141,935 ECGs.
Associations: age >65 y, AA race, prior Hx
VA, COPD, ST changes
QTc 300 msec assoc w increased
mortality: HR: 2.6 (95% CI: 1.93.7)
Guerrier K et al. Study type: Single Inclusion criteria: 1 endpoint: Prevalence short QTc 340 Short QTc 340 msec prevalence
Circ Arrh EP 2015 center retrospective Screened 272, 504 ECGs msec in patients <21 y old, deaths 0.05% in <21 y old
(414) <21 y for QTc340 msec
26386018 Size: Results: Prevalence 0.05%, 76% males Short QT rare, increased
Exclusion criteria: N/A Females shorter QTc 312 vs 323 msec, prevalence in males
p=0.03
2 deaths: respiratory; dilated
cardiomyopathy

227

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Bun SS et al. JCE Study type: case Inclusion criteria: 28 y old 1 endpoint: treatment electrical storm Case report efficacy of
2012 (415) report ACA while asleep, QTc 320 in short QTS isoproterenol in treating recurrent
22493951 msec, admitted with VF in short QT
Size: 1 electrical storm, 8 VF Results: isoproterenol infusion resulted
arrests while in sinus rhythm
sedated/hypothermia

Exclusion criteria: N/A

Dhutia H et al. Br J Study type: single Inclusion criteria: Healthy 1 endpoint: Prevalence and significance Males, Afro-Caribbean ethnicity
Sports Med 2016 center retrospective people ages 1435 y of short QTS among healthy young had strongest association with
(416) undergoing screening with individuals short QT
26400956 Size: screening hx, PE, ECG Short QTc 320 msec: excellent
18,825 patients Results: QTc 320 msec: 0.1%, 26 medium term prognosis in young
Exclusion criteria: N/A patients patients
QTc 330 msec: 0.2%, 44 Recommend using QTc 320
patients msec to prevent over-diagnosis
QTc <380 msec: 7.9%, 1478
patients
QTc <390 msec: 15.8%, 2973
patients
Followup 5.31.2 y, no deaths

Data Supplement 45. RCTs Related to VA in the Structurally Normal Heart (Section 8)
Study Acronym; Aim of Study; Patient Population Study Intervention (# Endpoint Results Relevant 2 Endpoint (if
Author; Study Type; patients) / (Absolute Event Rates, any);
Year Published Study Size (N) Study Comparator (# P values; OR or RR; & 95% Study Limitations;
patients) CI) Adverse Events
Ling et al. Aim: to compare the Inclusion criteria: Intervention: RF catheter 1 endpoint: The 1 end RF Catheter ablation is
2014 (417) efficacy of (1) ablation of RVOT point was recurrence of more effective than AAD
24523413 radiofrequency catheter frequent Comparator: RVOT VPBs at a rate of for treatment of frequent
ablation (RFCA) vs. symptomatic VPBs Antiarrhythmic premature beats arising
from the RVOT medications from the RVOT.
228

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
AAD for treatment of documented by 12- 300 beats per day
patients with frequent lead documented by 24 h
ventricular premature ECG to have Holter monitoring. The 2
beats (VPBs) originating inferior axis and left variables of interest
from the right bundle-branch including the number of
ventricular outflow tract block (LBBB) QRS VPBs, the burden
(RVOT). morphology of VPBs (the number of
(2) >6000 VPBs per VPBs/ total QRS
Study type: 24h on Holter complexes100%), and
Prospective, RCT monitoring. LVEF at each follow-up
time point were collected
Size: 330 patients Exclusion criteria:
(1) the presence of During the 1y follow-up
non-RVOT period, VPB
origin for VPBs recurrence was
indicated by an S significantly lower in
wave in lead I, R- patients randomized to
wave duration RFCA group (32 patients,
index in V1 and 19.4%) vs. AAD group (146
V20.5, and R/S patients, 88.6%; p<0.001,
wave amplitude log-rank test). In a Poisson
index in V1 and generalized estimating
V20.311; equations regression
(2) previous AAD model, RFCA
therapy; was associated with a
(3) evidence of any greater decrease in the
structural burden of VPBs (incidence
heart disease; rate ratio: 0.105; 95% CI:
(4) hyperthyroidism 0.1040.105; p<0.001)
or electrolyte compared with AAD. In a
disturbance; liner GEE model, the LVEF
(5) drug had a tendency
toxicity; to increase after the
(6) diabetes treatment in both groups
mellitus; (coefficient, 0.584; 95% CI:
(7) BP>165/100 mm 0.4670.702; p<0.001).
Hg;

229

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
(8) significant
impairment of renal
function;
(9) QT interval>450
ms in the absence
of bundle-branch
block;
(10) significant AV
conduction disease
and left or right
bundle-branch
block
Krittayaphong Study type: Inclusion criteria: Intervention: 1 endpoint: BB may be useful for
et al. 2002 (94) RCT VA with LBBB, Atenolol 50-100mg/day Atenolol significantly patients with RVOT and
12486439 inferior axis decreased PVC count symptomatic VA.
Aim: To determine the morphology. Comparator: Placebo (p=0.001) and average
efficacy of atenolol in Symptomatic (VA heart rate (p<0.001)
the treatment of disturbed their compared to placebo.
symptomatic VA from daily activities) Both placebo and atenolol
RVOT compared with decreased symptom
placebo Exclusion criteria frequency.
SHD.
Size: 52

Data Supplement 46. Nonrandomized Trials, Observational Studies, and/or Registries Related to Outflow Tract and AV Annular VA
(Section 8.1)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Liao et al. 2015 (418) Study type: Inclusion criteria: Results: Right ventricular outflow tract VAs may
26670064 Single Center Patients with idiopathic Among 244 patients with require ablation within the pulmonic
Observational VAs that were LBBB and inferior QRS axis valve sinus cusps.
successfully ablated VAs, 24 patients required
Size: within the pulmonic ablation within the pulmonic
24 patients valve sinus cusps sinus cusps.
230

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Successful ablation within the
Exclusion criteria: none right PV sinus in 10 patients,
the left sinus in 8, and
anterior sinus in 6.
There were no complications.
Morady et al. 1990 Study type: Single Inclusion criteria: Results: DC shock ablation in RVOT VT can be successfully ablated
(419) Center observational Consecutive patients the RVOT rendered 9 of 10 with DC shock ablation with high efficacy
2242533 undergoing DC Shock patients free of VT over a and low complications.
Size: 10 patients catheter ablation of mean follow-up of 33+18 mo.
RVOT VT There were no complications.

Exclusion criteria: none


Yamada et al. 2008 Study type: Single Inclusion criteria: Results: The aortic valve sinuses are a common
(420) Center Observational Idiopathic VAs Left coronary cusp in 24 location of outflow tract arrhythmias that
18598894 undergoing catheter patients (54.5%), can be effectively and safely ablated with
Size: 265 patients ablation Right coronary cusp in 14 RF current.
patients (31.8%),
44 patients with VAs Right-Left cusp junction in 5
mapped and ablated patients (11.4%), and
within the aortic sinuses Noncoronary cusp in 1 pt.

Successful catheter ablation in


44/44 patients (100%).
No complications.

Yamada et al. 2010 Study type: Inclusion criteria: Results: LV summit VAs may be ablated within
(421) Single Center Among 221 consecutive Successful ablation from the the GCV or inferior to the GCV on the
20855374 Observational patients with LV Great Cardiac Vein in 14 epicardial surface, though sites superior
Idiopathic VAs, 27 patients and on the epicardial to the GCV are often inaccessible to
Size: 27 patients patients had VAs mapped surface of the LV in 4. ablation.
and ablated on the In 5 patients ablation
Summit of the LV abandoned because of origin
in the inaccessible region. In 4
Exclusion criteria: N/A patients ablation abandoned
due to close proximity to
epicardial coronary artery.

231

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Mountantonakis et al. Study type: Single Inclusion criteria: Results: Although ablation at the earliest CVS
2010 (422) Center Observational Among 511 consecutive Twenty-five (53%) were in the site is effective, it is often (62%)
20855374 patients with non-scar great cardiac vein, 19 (40%) in precluded, mainly because of proximity
Size: 47 patients related VAs, 47 patients the anterior interventricular to coronary arteries. Ablation at adjacent
were found to have a site vein, and 3(7%) in the middle CVS and non-CVS sites can be successful
of origin within the cardiac vein. in 55% of these anatomically challenging
Coronary Venous System Successful ablation achieved cases, for an overall ablation success rate
(CVS). in 17 of 18 (94%) ablated at of 70%.
the earliest CVS site and in 16
Exclusion criteria: N/A of 29 (55%) ablated at
adjacent CVS or non-CVS sites.
Doppalapudi et al. Study type: Inclusion criteria: Results: Idiopathic VT may arise by a focal
2009 (423) Single Center Among 340 patients with VT was sustained and rapid mechanism from the epicardium at the
19121799 Observational idiopathic VT referred for (mean cycle length 264 msec) crux in close proximity to the posterior
ablation, four were in all patients and was descending coronary artery. This
Size: 4 patients identified with VT that associated with syncope or syndrome can result in rapid,
was mapped to the presyncope in three. VT was catecholamine-sensitive VT and requires
epicardium at the crux. induced with programmed careful attention to the posterior
stimulation or burst pacing in descending coronary artery during
Exclusion criteria: N/A all 4 patients but required ablation.
isoproterenol infusion in
three.
Konstantinidou et al. Study type: Inclusion criteria: Results: RVOT access is feasible with the
2011 (424) Single Center 13 patients presenting The RVOT was reached in all Magnetic Navigation System, while RVOT
21307021 Observational with VT suggestive of patients utilized solely with mapping and ablation appear to be safe,
RVOT origin with ablation the Magnetic Navigation fast, and effective.
Size: 13 patients guided by Magnetic System. Successful RVOT
Navigation ablation was achieved in (135)
(92.3%) patients.
Exclusion criteria: N/A No Complications occurred.
During a mean follow-up of
252+211 d, clinical
arrhythmia recurrence was
observed in 1 of 13 (7.7%)
patients.

232

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Ouyang et al. 2002 Study type: Inclusion criteria: Results: VAs may arise in either the right or left
(425) Single Center Consecutive patients The RVOT was site of origin in ventricular outflow tracts and can be
11823089 Observational with VAs from the right 7 patients and aortic sinuses safely ablated with RF current.
ventricular outflow tract in 8 patients.
Size: 15 patients or aortic sinuses The left coronary cusp was
the site of origin in 5 of 7
Exclusion criteria: N/A patients and the right
coronary cusp in 2 of 7
patients with aortic sinus VAs
Tada et al. 2005 (426) Study type: Single Inclusion criteria: Results: VAs may arise from the anterolateral,
15766824 Center Observational Consecutive patients Among 352 patients with posterior, and posteroseptal regions of
with VAs mapped to the idiopathic VAs, 19 (5%) had the mitral annulus and can be effectively
Size: 19 patients mitral valve annulus mitral annular VAs. and safely ablated with RF current.
11 (58%) originated from the
Exclusion criteria: N/A anterolateral mitral annulus,
2 from the posterior mitral
annulus, and 6 from the
posteroseptal mitral annulus.
Successful ablation achieved
in 19/19 patients (100%).
No complications observed.

Over a follow-up period of


21+15 mo, there were no
recurrences of VAs after
ablation.
Tada et al. 2008 (427) Study type: Single Inclusion criteria: Results: A site of origin in the Pulmonary artery
18313601 Center Observational Cases of VAs mapped Among 276 patients with VAs should be suspected when mapping and
and ablated within the referred for RF ablation, 12 ablation of apparent RVOT VAs is not
Size: 12 patients Pulmonary Artery. patients were identified with successful within the RVOT. Ablation
a successful site of catheter within the pulmonary artery is safe and
Exclusion criteria: N/A ablation within the pulmonary effectifve.
artery.

All 12 patients had attempted


ablation within the RVOT with

233

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
a change in the QRS
morphology after ablation.
A characteristic prepotential
was recorded within the
pulmonary artery in all
patients. Ablation was
successful within the
pulmonary artery in 12/12
patients (100%). There were
no complications.
No recurrences of VAs were
observed over a follow-up
period of 27+13 mo.
Tada et al. 2007 (428) Study type: Single Inclusion criteria: Results: Tricuspid annular VAs are not rare and
18313601 Center Observational Consecutive patients Among 454 consecutive ablation has a higher efficacy for freewall
with idiopathic VAs patients with idiopathic VAs, than septal sites.
Size: 38 patients mapped and ablated on 38 patients (8%) were found
the tricuspid annulus to originate from the tricuspid
annulus.
Exclusion criteria: N/A 28 (74%) originated from the
septal tricuspid annulus 10
(26%) from the freewall
portion of the annuls.
Catheter ablation eliminated
90% of freewall VAs but only
57% of septal tricuspid
annular VAs.
There were no complications.

Kamioka et al. 2015 Study type: Inclusion criteria: Results: LVOT VAs may arise above or below the
(429) Single Center Consecutive patients Twelve patients had VAs aortic valve. Prepotentials are recorded
25633492 Observational with LVOT Vas mapped in the Aortic cusps, at the site of successful ablation in the
Size: 34 patients and 22 patients had VAs majority of patients with origin within the
Exclusion criteria: N/A mapped below the Aortic aortic sinuses but are rarely recorded
valve. below the aortic valve.

234

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Pre-potentials recorded in
91% of Aortic Sinus VAs and
13% below the aortic valve.

VAs successfully ablated in


34/34 patients (100%)
Nagashima et al. Study type: Single Site Inclusion criteria: Results: Ablation within the GCV requires
2014 (430) observational 30 patients with VAs with Angiography in 27 patients careful attention to the proximity of
25110163 early activation within showed earliest GCV site coronary arteries with the potential for
Size: 30 patients the Great Cardiac Vein within 5 mm of a coronary coronary arterial injury.
(GCV). artery in 20 (74%).
Ablation was performed in the
Exclusion criteria: N/A GCV in 15 patients and
abolished VA in 8. Ablation
was attempted at adjacent
non-GCV sites in 19 patients
and abolished VA in 5 patients
(4 from the left ventricular
endocardium and 1 from the
left coronary cusp).

After a median of 2.8 mo, 13


patients remained free of VA.
Major complications occurred
in 4 patients, including
coronary injury requiring
stenting.
Yamada et al. 2015 Study type: Single Inclusion criteria: Results: LVOT VAs originating from intramural
(431) Center observational 64 consecutive patients Among 64 patients, 14 foci could usually be eliminated by
25637597 study with symptomatic patients were identified with sequential unipolar radiofrequency
idiopathic sustained VTs intramural foci between the ablation and sometimes required
Size: 64 patients (VTs) (N=14), NSVT endocardium and epicardium simultaneous ablation from both the
(N=15), or premature which required sequential or endocardial and epicardial sides.
ventricular contractions simultaneous irrigated
(PVCs) (N=35), which unipolar radiofrequency
presumed origins ablation from the endocardial
identified in the AMC, LV

235

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
summit, or intramural and epicardial sides for their
sites between the elimination.
endocardium and Simultaneous ablation was
epicardium. most likely to be required
when the distance between
Exclusion criteria: N/A the endocardial and epicardial
ablation sites was >8 mm and
the earliest local ventricular
activation time relative to the
QRS onset during the VAs was
<30 ms at both ablation sites.
Hai et al. 2015 (432) Study type: Single Inclusion criteria: Results: Specific identification and targeting of
25637597 Center observational All patients who Among 21 patients, PPs when ablating VAs at the AMC may
study underwent successful prepotentials (PPs) were improve procedural success.
catheter ablation of VAs found at the ablation sites
Size: 21 patients at the Aortomitral preceding the ventricular EGM
Continuity (AMS) during arrhythmias in 13
(61.9%) patients and during
Exclusion criteria: N/A sinus rhythm in 7 (53.8%)
patients.
VAs with PPs were associated
with a significantly higher
burden of premature
ventricular complexes (PVCs;
26.110.9% vs. 14.910.1%,
p=0.03), shorter ventricular
EGM to QRS intervals
(9.028.5 msec vs. 33.18.8
msec, p=0.03), lower pace
map scores (8.71.6 vs.
11.40.8, p=0.001), and a
trend toward shorter V-H
intervals during VA (32.1 8.6
msec vs. 76.311.1 msec,
p=0.06) as compared to those
without PP.

236

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Yamada et al. 2010 Study type: Single Inclusion criteria: Results: The MDI has limited value for
(433) Center observational All patients who 48 consecutive patients discriminating endocardial from
19804552 study underwent successful undergoing successful epicardial VA origins in sites adjacent to
catheter ablation of VAs catheter ablation of idiopathic the LSOV probably due to preferential
Size: 21 patients at the Aortomitral VAs originating from the left conduction, intramural VA origins or
Continuity (AMS) coronary cusp (LCC, N= 29), myocardium in contact
aortomitral continuity (AMC, with the LCC.
Exclusion criteria: N/A N=10) and great cardiac vein
or anterior interventricular
cardiac vein (Epi, N= 9).
An S wave in lead V5 or V6
occurred significantly more
often during both the VAs and
pacing from the AMC than
during that from the LCC and
Epi (p<0.05 vs. p=0.0001). For
discriminating whether VA
origins can be ablated
endocardially or epicardially,
the maximum deflection index
(MDI = the shortest time to
the maximum deflection in
any precordial lead/QRS
duration) was reliable for VAs
arising from the AMC (100%),
but was less reliable for LCC
(73%) and Epi (67%) VAs. In 3
(33%) of the Epi VAs, the site
of an excellent pace map was
located transmurally opposite
to the successful ablation site
(LCC = 1 and AMC = 2).

237

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 47. Nonrandomized Trials, Observational Studies, and/or Registries of Catheter Ablation in Papillary Muscle VA - (Section
8.2)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Doppalapudi et al. Study type: Single Inclusion criteria: Among 290 patients with Posteromedial papillary muscle VT is
2008 (434) Site Observational VT mapped to the idiopathic VAs, 7 were found catecholamine sensitive with a focal
19808390 Posterior Papillary to have origin in the mechanism that is amendable to
Size: 9 patients Muscle of the LV Posteromedial PM. catheter ablation. Catheter stability may
All patients had RBBB and be difficult and multiple RF applications
Exclusion criteria: none Superior QRS axis. are usually required.
No patient had SHD.
VT had focal mechanism,
sensitive to catecholamines

Results: Successful catheter


ablation in all patients without
complications.

Yamada et al. 2010 Study type: Single Inclusion criteria: Among 159 consecutive VT of focal origin may occur in either
(435) Site Observational VT mapped to the patients with idiopathic VAs the posteromedial of the anterolateral
20558848 Posteromedial or mapped to the LV, the site of PMs of the LV. Catheter ablation often
Size: 19 patients Anterolateral Papillary origin was in the requires multiple RF applications over a
Muscles of the LV Posteromedial PM in 12 and wide area suggesting an origin deep
the Anterolateral PM in 7. within the PM.
The recurrence risk after initially
Exclusion criteria: none Results: Successful ablation successful ablation is higher than for
was achieved in 19/19 many other forms of idiopathic VT.
patients. Multiple QRS
morphologies were observed
in 47% of patients and in 7
patients ablation on both
sides of the PM were
required. No complications
were observed.
Recurrence of PM VAs was
observed in 2/19 patients.

238

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Yokokawa et al. 2010 Study type: Single Inclusion criteria: Results VAs may originate in the papillary
(436) Site Observational VT mapped to the 40 consecutive patients muscles of both the LV and the RV. PVCs
20637311 Posteromedial or referred from the papillary muscles are often
Size: 40 patients anterolateral Papillary for ablation of symptomatic pleomorphic.
Muscles of the LV premature ventricular Catheter ablation is successful in over
complexes 80% of cases, with greater mass of the
Exclusion criteria: None (PVCs) (N=19) or VT (VT) papillary muscle predicting lower efficacy
(N=21) originating from a of ablation.
Papillary muscle in the LV
(N=32) or RV (N=8).

Antiarrhythmic drugs failed to


control the VAs in 24 patients.
20 of 40 patients (50%) had
SHD: prior MI in 10 patients,
dilated cardiomyopathy in 9,
and VHD in 1 pt.

Catheter ablation was acutely


successful in 33 of 40 patients
(83%).

Pleomorphic QRS
morphologies observed in
31/40 patients.
By MRI, the mass of the
arrhythmogenic PM was
greater in patients with failed
than successful ablations.
In follow-up, the PVC burden
was reduced from 15%+11%
to 3%+3%; p<0.01) after
successful ablation.
Crawford et al. 2010 Study type: Single Inclusion criteria: Results: PVCs and VT may originate in the RV
(437) Site observational A total of 15 distinct PAP VAs PAPs. Radiofrequency ablation is
20206325 was mapped to the posterior effective in eliminating these

239

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 8 patients VAs mapped to the (N=3), anterior (N=4), or arrhythmias with low risk of
papillary muscles in the septal (N=8). complications.
right ventricle.
Successful ablation achieved
Exclusion criteria: none in all 8 patients.
The PVC burden was reduced
from 17%+20% preablation to
0.6%+0.8% postablation.
Ban et al. 2013 (438) Study type: Single Inclusion criteria: Results: In PMVT, a high-amplitude, discrete
24385992 Site Observational Among 284 patients with Successful catheter ablation potential before the QRS and slow down-
idiopathic VAs was achieved in 7 of 8 (87.5%) stroke of the initial Q wave on the
Size: 12 patients undergoing ablation, 12 patients with high amplitude unipolar electrogram at ablation sites are
patients were identified electrograms at the earliest related to favorable outcome after RF
with VAs originating from site of origin. catheter ablation.
the Papillary Muscles of The 4 patients with low
the LV. amplitude and fractionated
electrograms had recurrences
of VAs after ablation.

The mean duration from


onset to peak downstroke (t)
on the unipolar electrogram
was significantly longer in the
successful group than in the
recurrence group (588 ms vs.
379 ms, p=0.04). A slow
downstroke >50 ms of the
initial Q wave on the unipolar
electrogram at ablation sites
was also significantly
associated with successful
outcome (85.7% vs. 25.0%,
p=0.03).

240

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 48. Nonrandomized Trials, Observational Studies, and/or Registries Related to Interfascicular Reentrant VT (Belhassen
Tachycardia)- (Section 8.3)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Nogami et al. 2000 Study type: Inclusion criteria: Results: Verapamil sensitive idiopathic LV VT is
(439) Multicenter 20 consecutive patients Sustained VT could be a reentrant tachycardia involving a
10987604 Observational with verapamil-sensitive induced by programmed discrete longitudinal pathway in the LV
left VT electrical stimulation, septum and retrograde conduction over
Size: 20 patients exhibiting a RBBB and entrained by rapid ventricular the His Purkinje network. Catheter
left-axis deviation QRS pacing, and terminated by ablation is highly successful with a low
who underwent RF verapamil in all patients. risk of complications.
ablation. Two discrete potentials could
be recorded on the LV septum
Exclusion criteria: with antegrade conduction
None (P1) and retrograde
conduction (P2).
RF current applied to the exit
site of P1 terminated VT in all
patients.
The interval between the LV
and the P1 potential
demonstrated decremental
conduction and verapamil
sensitivity.
Liu et al. 2015 (440) Study type: Inclusion criteria: Results: Ablation of FVT guided by activation
10987604 Single Center Consecutive patients 120 patients with idiopathic mapping is associated with a single
Observational with Idiopathic fascicular fascicular VT (mean age, procedural success rate of 80.3% without
VT undergoing catheter 29.312.7 y; 82% men; all the use of AAD.
Size: 120 patients ablation. with normal EF).
Catheter ablation acutely 23 patients (20%) developed new onset
Exclusion criteria: successful in 117 of 120 LPF block, whereas 67 patients (58.3%)
None patients. Over median follow- exhibited rightward shift in their frontal
up of 55.7 mo, VT recurred in axis compared with baseline.
17 patients, all successfully re- There were no complications from the
ablated. procedure.

241

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Lin et al. 2005 (441) Study type: Inclusion criteria: Results: A linear ablation lesion perpendicular
26386017 Single Center Consecutive patients Among 15 patients with to the long axis of the LV across the left
Observational with idiopathic fascicular idiopathic fascicular VT, 6 side of the interventricular septum is an
VT undergoing catheter (40%) had VT that was not effective ablation strategy for patients
Size: 15 patients ablation inducible with programmed with idiopathic fascicular VT that is non-
stimulation and isoproterenol. inducible.
Exclusion criteria: For these patients, a linear
N/A lesion was placed
perpendicular to the long axis
of the ventricle approximately
midway from the base to the
apex in the region of the mid
to mid-inferior septum.
Left posterior fascicular block
developed in 2 of 6 patients.
No spontaneous arrhythmias
occurred during follow-up to
168 mo (range 630 mo).

Data Supplement 49. Nonrandomized Trials, Observational Studies, and/or Registries Related to Idiopathic Polymorphic VT/VF - (Section 8.5)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Hassaguerre et Study type: Inclusion criteria: Results: Idiopathic VF is often triggered by short
al. 2002 (442) Multi-Center 16 patients with 16 patients with idiopathic VF coupled PVCs from the RVOT or the
11879868 Observational idiopathic VF treated with triggered by short coupled PVCs Purkinje system. The initiating focus can
catheter ablation (mean 300 msec). The mean PVC be successfully ablated with low risk of
Size: 16 patients frequency per day was 9618. complications.
Exclusion criteria: N/A The initiating focus was in the
RVOT in 4 patients, the RV
Purkinje in 4 patients, the LV
Purkinje in 7 patients, and both
the RV and LV Purkinje in 1 pt.

242

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Initially successful ablation of the
triggering PVC focus in 16/16
patients.
Long term freedom from VF
observed in 13 patients.
VALIANT Aim: To evaluate risk Inclusion criteria: Intervention: Analysis of rates of Each 5% lower LVEF was associated
Solomon et al. and predictors of SCD Patients with first or SCD. Evaluation of EF determined with a 21% increase in adjusted risk of
2005 (30) in patients post MI subsequent MI with HF, by echocardiography as well as SCD or CA with resuscitation.
15972864 with left ventricular LV dysfunction, or both other parameters.
dysfunction and/or HF
Exclusion criteria: ICD in Comparator: N/A
Study type: place prior to 1 endpoint: The risk of sudden
Observational study of randomization deathwas greatest in the first 30 d
patients enrolled in a after MI: 1.4% per mo, 95% CI:
RCT 1.2%1.6% and decreased to
0.14% per mo 95% CI: 0.11%
Size: 14,609 patients 0.18% after 2 y after MI. Patients
with LVEF <30% were at the
greatest risk for SCD

Linzer et al. 1990 Study type: Inclusion criteria: 1 endpoint: Monitor up to 1mo 25% yield for syncope Dx after negative
(25) observational Syncope with negative with Loop Holter
2371954 Holter VT/VF uncommon (1 pt)
Size: 57 Results: arrhythmia was the
Exclusion criteria: cause of symptoms (diagnostic
Patients who had yield 25%; 95% CI: 1438%).
undergone VT (1 patient), high grade AV
electrophysiology study block
(2 patients), supraventricular
tachycardia (1 patient),
asystole or junctional bradycardia
from neutrally mediated syncope
(3 patients) and normal cardiac
rhythms (the remaining 7
patients).
Noda et al. 2005 Study type: Single Inclusion criteria: Results: PVCs from the RVOT may trigger VF
(443) Center Observational when the coupling interval is short (<320

243

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
16198845 16 patients who had Holter monitoring showed msec). The long term outcome after
Size: 16 patients documented VF or frequent PVCs with LBBB inferior ablation of the triggering focus is
syncope out of a total of QRS axis with mean coupling excellent.
101 patients with RVOT interval of 245+ 28 msec.
VAs undergoing catheter RF ablation targeting the initiating
ablation PVC focus acutely successful in
16/16 patients.
Over mean follow-up period of
5439 mo, no recurrences of
syncope or VF.

Haissaguerre et Study type: Inclusion criteria: Results: Idiopathic VF can be successfully


al. 2002 (444) Multicenter 27 patients undergoing Premature beats were elicited ablated by targeting the initiating focus
12186801 Observational catheter ablation of from the Purkinje conducting which is usually in the Purkinje system or
Size: 27 patients idiopathic VF without system in 23 patients: from the RVOT.
SHD left ventricular septum in 10,
from the anterior right ventricle in
9, and from both in 4, and from
the RVOT in 4 patients.
The interval from the Purkinje
potential to the following
myocardial activation varied from
10150 ms during premature beat
but was 115 ms during sinus
rhythm, indicating location at
peripheral Purkinje arborization.
The accuracy of mapping was
confirmed by acute elimination of
premature beats during local
radiofrequency delivery. During a
follow-up of 2428 mo, 24
patients (89%) had no recurrence
of VF without drug
Van Herendael et Study type: Single Inclusion criteria: Results: Catheter ablation of VPD-triggered
al. 2014 (445) Center Observational 30 patients from among In 21 patients, VF/PMVT occurred VF/PMVT is highly successful. Left
24398086 1132 consecutive in the setting of cardiomyopathy; ventricular outflow tract and papillary
Size: 30 patients patients undergoing in 9 patients, VF/PMVT was muscles are common and are previously

244

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
catheter ablation of VAs idiopathic. The origin of VPD unrecognized sites of origin of these
of all types trigger was from the Purkinje triggers in patients with and without SHD.
network in 9, papillary muscles in
8, left ventricular outflow tract in
9, and other low-voltage areas
unrelated to Purkinje activity in 4.
Acute VPD elimination was
achieved in 26 patients (87%),
with a decrease in VPDs in
another 3 patients (97%).
During median follow-up of 418 d
(interquartile range [IQR] 144-
866), 5 patients developed a
VF/PMVT recurrence after a
median of 34 d.
Sadek et al. 2015 Study type: Inclusion criteria: Results: VAs originating from the moderator
(446) Single Center 10 patients with VAs VF was the clinical arrhythmia in 7 band may present with VF. Catheter
25240695 Observationa. mapped to moderator patients and monomorphic VT in 3 ablation is effective, though the risk of
Size: 10 patients band in the RV patients. requiring more than one procedure may
undergoing catheter be higher than for other sites.
ablation Six patients required a repeat
procedure.
After mean follow-up of 21.511.6
mo, all patients were free of
sustained VAs, with only 1 patient
requiring AAD therapy and 1
patient having isolated PVCs no
longer inducing VF. There were no
procedural complications.
Tester DJ et al. Study type: Inclusion criteria: 1 endpoint: genetic mutation Recommend genetic screening for
Mayo Clinic Proc retrospective single Unexplained drowning yield in unexplained drowning unexplained drowning, especially if
2011 (447) center patients 1988-2010 victims positive family Hx of drowning, prolonged
21964171 molecular autopsy, mean QTc
Size: 35 age 1712 y (4-69 y). 28 Results: 23% positive mutations,
swimming (age 15.7 y), 7 8/28 swimming, 0/7 bathtub
bathtub (age 23 y). PCR Pos family Hx 43%: syncope,
seizures, CA, near-drowning or

245

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
DNA sequencing for LQTS drowning. Among 11 patients with
1-3, RYR2 positive personal or family hx,
Exclusion criteria: N/A 64% gene positive
N/A

Tzimas I et al. Int Study type: Inclusion criteria: 1 endpoint: Testing mutations in NOS1AP mutation of KCNq1 may be
J Legal Med 2016 retrospective Genotyping performed in 19 variants in drowning/water significant in drowning victims.
(448) corpses found in water: related deaths. Recommend molecular autopsy in
27460199 Size: 171 drowning, unclear deaths. unexplained water deaths.
Results: one SNP of KCNQ1 noted
Exclusion criteria: N/A NOS1AP significance
Anderson JH et Study type: Inclusion criteria: 1 endpoint: yield of genetic In decedents with exertion related SUD
al. Circ CV Gen retrospective single Exertion related SUDY testing in decedents with exercise <20 y, overall yield 44%,
2016 (449) center decedents (sudden related sudden death Yield higher in probands <11 y.
27114410 unexplained death in
Size: 32 young) Results: PCR DNA testing putative
ages 1-19 y mutation in 34% (11 patients,
Mean age 115 y Family LQTS, CPVT).
Hx SCD age <50 y in 10% Subsequent WES performed in 21
patients, yield 3/21, 14%
Molecular autopsy 1998- (calmodulin 2, PKP2 1-ARVC).
2010. Calmodulin deaths 2, 5 y.
DNA sequencing (PCR)
followed by whole-exome Yield higher among decedents
sequencing aged 110 y (91%) vs. 1119 y
(19%), p=0.0001
Exclusion criteria: N/A
Wang D et al. Study type: Inclusion criteria: SUD 1 endpoint: Yield of Overall genetic testing positive in
Forensic Sci Int Retrospective cohort channelopathy genetic channelopathy genetic screening 13.5%19.5% of autopsy negative sudden
2014 (450) testing in NYC 2008-2012. in ethnically diverse population of death
24631775 Size: 274 LQTS, RYR2 testing. SUCD Genetic testing information should be
Ages 1 y, 141 patients, provided to the family members with
51%, Results: Gene positive: 13.5% proper counseling along with the choices
Age 158 y, 133 cases, infants, 19.5% older of further clinical evaluation

246

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
African Americans 48%, SCN5A positive, 68% infants, 50%
Hispanic 22%, Caucasian non-infants
16% AA carried more SCN5A, KCNQ1
Exclusion criteria: variants vs other ethnic groups;
autopsy positive Whites: more RYR2
LQTS more prevalent during sleep
related deaths, RYR2 active
Kumar S et al. Study type: Inclusion criteria: 1 endpoint: Evaluate yield of Clinical + targeted genetics yield: SADS:
Heart Rhythm Autopsy negative sudden comprehensive evaluation of 18%, UCA 62%
2013 (451) Size: 502 unexplained death SADS and UCA Inherited cardiac disease diagnosed
23973953 syndrome (SADS) and only in families with multiple events
unexplained CA (UCA) Results: SADS: yield 18%; LQTS in Recommend ongoing periodic clinical
(patients resuscitated young 20 y; Brugada in age 40 evaluation of children/young family
successfully), mean age y. members for developing disease
32 y. UCA: yield 62%: mainly LQTS and
Clinical evaluation (ECG, BrS; CPVT, ER, ARVC, Short QT.
EST, echo) w targeted Targeted genetic tesing in patients
genetic testing. SADS with proven or suspected
mean age 24 y, UCA 32 y. phenotoype: molecular dx SADS
Exclusion criteria: N/A 35%, UCA 48%.

Data Supplement 50. Nonrandomized Trials, Observational Studies, and/or Registries of PVC-induced Cardiomyopathy - (Section 9)
Study Study Patient 1 Endpoint and Results Summary/Conclusion
Acronym; Type/Design; Population (P values; OR or RR; Comment(s)
Author; Study Size & 95% CI)
Year
Published
Ban et al. Study type: Inclusion criteria: Results: A PVC burden >26%/d
2013 (452) Single Site PVC burden >10% Left ventricular dysfunction (EF predicts LV dysfunction
23194696 Observational per 24 h and no <50%) was present in 28 of 127 with sensitivity of 70%
known SHD patients (22.0%). The mean PVC and specificity of 78%.
Size: 127 burden (31+11 vs. 22+10%, Thus, PVC induced LV
patients p<0.001), the presence of non- dysfunction is reversible

247

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion sustained VT (53.6 vs. 33.3%, with catheter ablation
criteria: SHD p<0.05), and the presence of a though there is wide
retrograde P-wave following a variability in the PVC
PVC (64.3 vs. 30.3%, p=0.001) burden associated with
were signicantly greater in those reduced LVEF.
with LV dysfunction than in those
with normal LV function. The cut-
off PVC burden related to LV
dysfunction was 26%/day, with a
sensitivity of 70% and a specicity
of 78%.
The origin sites of PVCs, the acute
success rate, and the recurrence
rate during follow-up after RFCA
were similar. In a multivariate
analysis, the PVC burden (OR:
2.94; 95% CI: 0.903.19, p=0.006)
and the presence of retrograde P-
waves (OR: 2.79; 95% CI: 1.08
7.19, p=0.034) were
independently associated with
PVC-mediated LV dysfunction.
Study type: Inclusion criteria: Results: Idiopathic VF is often
Hassaguerre Multi-Center 16 patients with 16 patients with idiopathic VF triggered by short
et al. 2002 Observational idiopathic VF triggered by short coupled PVCs coupled PVCs from the
(442) treated with (mean 300 msec). The mean PVC RVOT or the Purkinje
11879868 Size: 16 catheter ablation frequency per day was 9618. system. The initiating
patients The initiating focus was in the focus can be successfully
Exclusion RVOT in 4 patients, the RV ablated with low risk of
criteria: N/A Purkinje in 4 patients, the LV complications.
Purkinje in 7 patients, and both
the RV and LV Purkinje in 1 pt.
Initially successful ablation of the
triggering PVC focus in 16/16
patients.
Long term freedom from VF
observed in 13 patients.

248

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: Inclusion criteria: Results: Idiopathic VF can be
Haissaguerre Multicenter 27 patients Premature beats were elicited successfully ablated by
et al. 2002 Observational undergoing from the Purkinje conducting targeting the initiating
(444) Size: 27 catheter ablation system in 23 patients: from the focus which is usually in
12186801 patients of idiopathic VF left ventricular septum in 10, the Purkinje system or
without SHD from the anterior right ventricle RVOT.
in 9, and from both in 4, and from
the RVOT in 4 patients.
The interval from the Purkinje
potential to the following
myocardial activation varied from
10150 ms during premature
beat but was 115 ms during
sinus rhythm, indicating location
at peripheral Purkinje
arborization.
The accuracy of mapping was
confirmed by acute elimination of
premature beats during local
radiofrequency delivery. During a
follow-up of 2428 mo, 24
patients (89%) had no recurrence
of VF without drug
Lee et al. Study type: Inclusion criteria: 1 endpoint: All cause mortality ICD was not associated
2015 (453) Single Center, Continuous Flow with improved survival.
25940215 Retrospective LVAD only Results:
review, 2004 64 patients. Had ICDs.
2013 Exclusion Death occurred in 15 (38%)
criteria: N/A patients in the no ICD group vs.
Size: 100 18 (30%) in the ICD group.
Univariate analysis demonstrated
a marginal early survival benefit
at up to 1 y. No difference after 1
y.
Multivariate analysis did not
show any significant predictor of
survival.
249

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
No patients died of SCD.
Carballeira Study type: Inclusion criteria: Results: A QRS duration >153
Pol et al. Single Site Consecutive Of the 45 patients studied, 28 msec of high frequency
2014 (454) Observational patients without patients (62%) developed PVC- PVCs and a non-outflow
24184787 SHD who had related LV dysfunction and 17 tract site of origin are
Size: 45 >10% PVCs/d and patients (38%) remained with predictors of developing
patients normal LVEF normal LV function. PVC-induced LV
(>0.55) who were The PVC burden was similar dysfunction.
observed. (26.5% vs 26%) between the two
groups (p=NS).
Exclusion The QRS duration was
criteria: significantly greater for those who
Structural Heart developed LV dysfunction than
Disease those who did not (159 vs 142
msec, p<0.001).
A PVC QRS duration >153 msec
best predicted the development
of LV dysfunction (sensitivity 82%
and specificity 75%).
A non-outflow tract site of origin
was also an independent
predictor of LV dysfunction.
Deyell et al. Study type: Inclusion criteria: Results: For patients with a PVC
2012 (455) Single Center 114 consecutive Over a median follow-up of 10.6 burden >10%/d, LV
22640894 observational patients with PVC mo, 24 of 48 patients with LV dysfunction may reverse
burden >10%/d dysfunction were classified as after successful catheter
Size: 114 undergoing reversible and 13 of 48 as ablation. The more
patients catheter ablation. irreversible and 11 of 44 were prolonged the QRS
66 patients had excluded due to failed ablation. duration of the PVC the
preserved LV higher the risk that LV
function and 48 There was a gradient of VPD QRS dysfunction will not
patients had duration between the control, improve.
impaired LV reversible, and irreversible groups
function (mean VPD QRS 135, 158, and 173
ms, respectively; p<0.001). This
Exclusion gradient persisted even for the
criteria: same site of origin. In multivariate

250

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Structural Heart analysis, the only independent
Disease predictor of irreversible LV
function was VPD QRS duration
OR: 5.07; 95% CI: 1.2221.01 per
10-ms increase).
Del Carpio Study type: Inclusion criteria: Results: A higher PVC burden
Munoz et al. Single Center 70 patients Patients with reduced LVEF and prolonged QRS
2011(456) Observational undergoing PVC (N=17) as compared to normal duration during PVCs
21332870 ablation without LVEF (N=53) had an increased may predict patients with
Size: 70 SHD. burden of PVCs (29.314.6% vs reversible, PVC-induced
patients Exclusion 16.713.7%, p=0.004), higher CM.
criteria: prevalence of NSVT (VT) [13 (76%)
Known SHD vs 21 (40%), p=0.01], longer PVC
duration (154.322.9 vs
145.620.8 ms, p=0.03) and
higher prevalence of multiform
PVCs [15 (88%) vs 31 (58%),
p=0.04].
There was no significant
difference in prevalence of
sustained VT, QRS duration of
normally conducted complexes,
PVC coupling interval, or delay in
PVC intrinsicoid deflection.
Olgun et al. Study type: Inclusion criteria: Results: The presence of
2011 (457) Single Center 51 consecutive Fourteen of the 21 patients (67%) interpolated PVCs was
21376837 Observational patients with with cardiomyopathy had predictive of the
PVCs undergoing interpolated PVCs, compared with presence of PVC -related
Size: 51 24 h Ambulatory only 6 of 30 patients (20%) cardiomyopathy.
patients Monitoring, without PVC-induced Interpolation may play an
including 21 cardiomyopathy (p<0.001). important role in the
patients with Patients with interpolated PVCs generation of PVC-
PVC-induced had a higher PVC burden than induced cardiomyopathy.
cardiomyopathy patients without interpolation
and 30 patients (28%12% vs. 15%15%;
without p=0.002). The burden of
cardiomyopathy. interpolated PVCs correlated with

251

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
the presence of PVC
Exclusion cardiomyopathy (21%30% vs.
criteria: 4%13%; p=0.008). Both PVC
Structural Heart burden and interpolation
Disease independently predicted PVC-
induced cardiomyopathy (OR:
1.07; 95% CI: 1.011.13, p=0.02;
and OR: 4.43; 95% CI: 1.0618.48,
p=0.04, respectively). The
presence of ventriculoatrial block
at a ventricular pacing cycle
length of 600 ms correlated with
the presence of interpolation
(p=0.004). Patients with
interpolation had a longer mean
ventriculoatrial block cycle length
than patients without
interpolated PVCs (520110 ms
vs. 39492 ms; p=0.01).
Hasdemir Study type: Inclusion criteria: Results: TICMP was relatively
et al. 2011 Single Center Seventeen of 247 Patients with TICMP compared to common (1 in every 15
(458) Observational patients with patients with preserved LVEF patients) in our study
21235667 PVCs (6.8%) who were more likely to be male (65% population. The
Size: 247 had Ambulatory vs 39%, p=0.043) and predictors of TICMP were
patients monitoring and asymptomatic (29% vs 9%, male gender, absence of
ECHO had p=0.018), and were more likely to symptoms, PVC burden
tachycardia have higher PVC burden (29.49.2 of 16%, persistence of
induced vs 8.17.4, p<0.001), persistence PVCs throughout the day,
cardiomyopathy of PVCs throughout the day (65% and the presence of
(TICMP) vs 22%, p=0.001), and repetitive repetitive monomorphic
monomorphic VT (24% vs 0.9%, VT
Exclusion p<0.001). PVC burden of 16% by
criteria: ROC curve analysis best separated
Structural Heart the patients with TICMP
Disease compared to patients with
preserved LVEF (sensitivity 100%,

252

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
specificity 87%, area under curve
0.96).
Baman et Study type: Inclusion criteria: Results: A PVC burden of >24%
al. 2010 (459) Single Center Consecutive A reduced LVEF (mean 0.370.10) was independently
20348027 Observational group of 174 was present in 57 of 174 patients associated with PVC-
patients referred (33%). Patients with a decreased induced cardiomyopathy.
Size: 174 for ablation of EF had a mean PVC burden of
patients frequent 33%13% as compared with those
idiopathic PVCs with normal left ventricular
function 13%12% (p<0.0001). A
Exclusion PVC burden of >24% best
criteria: separated the patient population
Structural Heart with impaired as compared with
Disease preserved left ventricular function
(sensitivity 79%, specificity 78%,
area under curve 0.89) The lowest
PVC burden resulting in a
reversible cardiomyopathy was
10%.
Kanei et al. Study type: Inclusion criteria: Results: A new index, which
2008 (460) Single Center Consecutive 24 patients had <1000 PVCs/24 h, incorporates PVC burden,
20348027 Observational group of 108 55 patients had 100010,000 QRS width and presence
patients referred PVCs/24 h, and 29 patients had of SHD or suspected EPI
Size: 108 for evaluation of 10,000 PVCs/24 h. The origin that best predicted
patients frequent prevalence of LV dysfunction was PVC-CMP.
idiopathic PVCs 4%, 12%, and 34%, respectively
from the RVOT (p=0.02). With logistic regression
analysis, non-sustained VT was an
Exclusion independent predictor of LV
criteria: dysfunction with OR: 3.6; 95% CI:
Structural Heart 1.310.1).
Disease

Hamon et Study type: Inclusion criteria: Results: LV dysfunction in the


al. 2016 (461) Single Center 107 consecutive Patients with decreased LV setting of frequent,
26924618 Observational patients (69 men; function had a greater PVC idiopathic PVCs may
mean age = burden on a 24-hour Holter represent a form of

253

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 107 5616 y) with monitor than patients with cardiomyopathy that can
patients frequent PVC normal EF (37%13% vs. be reversed by catheter
(23.111.5%) 11%10% of all QRS complexes; ablation of the PVCs.
referred for PVC p<0.0001). There was a significant
ablation. inverse correlation between the
PVC burden and the EF before
Exclusion ablation (r=0.73, p<0.0001).
criteria: PVCs originated in the right
Structural Heart ventricular outflow tract in 31
Disease (52%) of 60 patients, the LV
outflow tract in 9 (15%) of 60
patients, and in other sites in 13
(22%) of 60 patients. The site of
PVC origin could not be
determined in seven patients.
Ablation was completely
successful in 48 (80%) patients. In
patients with an abnormal EF
before ablation, LV function
normalized in 18 (82%) of 22
patients from a baseline of 34% to
59%7% (p<0.0001) within 6 mo.
In the 4 patients in whom
ablation was ineffective, the EF
further declined from 34%10%
to 25%7% (p=0.06) during
follow-up. In a control group of 11
patients with a similar PVC
burden (30%8%) and a reduced
EF (28%13%) who did not
undergo ablation, the EF
remained unchanged in 10/11
patients over 1917 mo of follow-
up and one patient underwent
heart transplantation.

254

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Bogun et al. Study type: Inclusion criteria: Results: LV dysfunction in the
2007 (462) Single Center 60 consecutive Patients with decreased LV setting of frequent,
17599667 Observational patients with function had a greater PVC idiopathic PVCs may
idiopathic, burden on a 24 h Holter monitor represent a form of
Size: 60 frequent PVCs than patients with normal EF cardiomyopathy that can
patients (>10/h), a (37%13% vs. 11%10% of all QRS be reversed by catheter
reduced LV EF complexes; p<0.0001). There was ablation of the PVCs
(EF; mean a significant inverse correlation
34%13%) was between the PVC burden and the
present in 22 EF before ablation (r=0.73,
(37%) patients p<0.0001).
PVCs originated in the right
Exclusion ventricular outflow tract in 31
criteria: (52%) of 60 patients, the LV
Structural Heart outflow tract in 9 (15%) of 60
Disease patients, and in other sites in 13
(22%) of 60 patients. The site of
PVC origin could not be
determined in seven patients.
Ablation was completely
successful in 48 (80%) patients. In
patients with an abnormal EF
before ablation, LV function
normalized in 18 (82%) of 22
patients from a baseline of 34% to
59%7% (p<0.0001) within 6 mo.
In the 4 patients in whom
ablation was ineffective, the EF
further declined from 34%10%
to 25%7% (p=0.06) during
follow-up. In a control group of 11
patients with a similar PVC
burden (30%8%) and a reduced
EF (28%13%) who did not
undergo ablation, the EF
remained unchanged in 10/11

255

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
patients over 1917 mo of follow-
up

Zhong et al. Study Type: Inclusion Criteria: Results: RFA appears to be


2014 (463) Single Center 510 patients with Of 510 patients identified, 215 more effective than AAD
24157533 Prospective frequent PVCs (40%) underwent RFA and 295 in PVC reduction and
observational (>1000/24 h) (60%) received AAD. The LVEF normalization
were treated reduction in PVC frequency was
Size: either by RFA or greater by RFA than with AAD (-
510 patients with AAD from 21,799/24 h vs -8,376/24 h;
January 2005 p<0.001). The LVEF was increased
through significantly after RFA (53%56%;
December 2010. p<0.001) but not after AAD (52%
Data from 24 h 52%; p=0.6) therapy. Of 121
Holter monitoring (24%) patients with reduced LVEF,
and 39 (32%) had LVEF normalization
echocardiography 50%. LVEF was restored in 25 of
before and 612 53 (47%) patients in the RFA
mo after group compared with 14 of 68
treatment were (21%) patients in the AAD group
compared (p=0.003). PVC coupling interval
between the less than 450 ms, less impaired
treatment 2 left ventricular function, and RFA
groups were independent predictors of
LVEF normalization performed by
Exclusion using multivariate analysis.
criteria:
Structural Heart
Disease

Kawamura Study type: Inclusion criteria: Results: In addition to the PVC


et al. 2014 Single Center 214 patients Among these patients, 51 (24%) burden, the CI-dispersion
(464) Observational undergoing had reduced LVEF and 163 (76%) and BMI are associated
24157533 successful had normal LV function. Patients with PVC-induced
Size: 214 ablation of PVCs with LV dysfunction had cardiomyopathy
patients who had no other significantly longer coupling
interval (CI) dispersion

256

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
causes of (maximum-CI-minimum-CI) and
cardiomyopathy had significantly higher PVC
burden compared to those with
Exclusion normal LV function (CI-dispersion:
criteria: 11525 msec vs. 9419 msec;
Structural Heart p<0.001; PVC burden: 19% vs.
Disease 15%; p=0.04). Furthermore,
patients with LV dysfunction had
significantly higher body mass
index (BMI) compared to those
with normal LV function (BMI>30
kg/m2; 37% vs. 13%; p=0.001).
Logistic regression analysis
showed that CI-dispersion, PVC
burden, and BMI (>30 kg/m2) are
independent predictors of PVC-
induced cardiomyopathy.
Yokokawa Study Type: Inclusion Criteria: Results: PVC-induced
et al. 2013 Single Center A consecutive The majority of patients (51 of 75, cardiomyopathy resolves
(465) observational series of 264 68%) with PVC-induced LV within 4 mo of successful
24612052 Size: patients with dysfunction had a recovery of LV ablation in most patients.
264 patients frequent function within 4 mo. In 24 (32%) In about one-third of the
idiopathic PVCs patients, recovery of LV function patients, recovery is
referred for PVC took more than 4 mo (mean 129 delayed and can take up
ablation, mo; range 5-45 mo). An epicardial to 45 mo. An epicardial
including 87 with origin of PVCs was more often origin predicts delayed
LV dysfunction present (13 of 24, 54%) in recovery of LV function.
patients with delayed recovery of
Exclusion LV function than in patients with
criteria: early recovery of LV function (2 of
Structural Heart 51, 4%; p<0.0001). The PVC-QRS
Disease width was significantly longer in
patients with delayed recovery
than in patients with recovery
within 4 mo (17021 ms vs
15916 ms; p=0.02). In
multivariate analysis, only an

257

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
epicardial PVC origin was
predictive of delayed recovery of
LV function in patients with PVC-
induced cardiomyopathy

Data Supplement 51. Nonrandomized Trials, Observational Studies, and/or Registries Related to Pregnancy - (Section 10.2)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Jeejeebhoy et al. Study type: Inclusion criteria: 1 endpoint: Both this Scientific Statement on
2015(466) Scientific Statement Comprehensive review N/A Cardiac Arrest in Pregnancy and the 2015
26443610 of the AHA and recommendations American Heart Association Guidelines
for management of CA Results: Update for Cardiopulmonary
Size: during pregnancy Specific recommendation for Resuscitation and Emergency
N/A management of CA during late Cardiovascular Care; Part 10: Special
Exclusion criteria: pregnancy and delivery. There are 2 Circumstances of Resuscitation,
N/A of major importance that are given recommend that in CA when the uterus is
the force of Recommendations in the above the umbilicus, left uterine
absence of supporting data on displacement (142) should be performed
outcomes (LOE-C): Left Uterine to relieve aortocaval compression during
Displacement during CPR when the CPR. While there is limited data on the
uterus is above the umbilicus; and relief of aortocaval compression by this
the 4-5 min rule for emergency C- maneuver, there is no data on the effect
section during CA PMCD. of LUD on outcomes. This is a Class I
Recommendation, with LOE C.
There is no specific data to support
these recommendations from the point of
view of outcomes yet they are woven in
to two recommendation documents
recently released.
The 4-5 min window for PMCD is also
based on limited theoretic information,
but does not have any scientific basis
supporting improved maternal or fetal
outcomes. It is a Class IIa
258

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
recommendation, LOE C. It is led to the
recommendation that a scalpel be
available for response teams on the
obstetrical units, and a recommendation
against moving the patient to operating
room or delivery suite, but rather doing
the PMCD on site.
Creagna A A, et al Study type: Inclusion criteria: 1 endpoint: Deaths during or within Pregnancy-related mortality ratios are
2014 (467) Analysis of De-identified maternal 1 y after pregnancy, with causes 34 times higher among black than white
3880915 surveillance data and related fetal based upon death certificate data. women
accumulated by deaths reported to CDC The data do not distinguish CA from
CDC (Division of by 52 voluntary Results: Pregnancy-related mortality other mechanisms of CV death; nor do
Reproductive reporting areas (50U.S. ratio increased steadily from 7.2 they distinguish tachyarrhythmic CA from
Health) states, New York City, deaths/100,000 live births in 1987 to other mechanisms.
and District of 17.8 deaths/100,000 live births in
Size: Columbia); based upon 2009. The reasons for this increase
Absolute numbers death certificate data are unclear.
not specified
Exclusion criteria: In parallel with this, there has been a
None specified decline in the contribution of the
traditional causes of pregnancy-
related mortality (i.e., hemorrhage,
sepsis, hypertensive disorders of
pregnancy), and the emergence of CV
and other medical conditions as
important contributors to mortality.
For the most recent surveillance
period shown (20062009), CV
conditions alone accounted for over
1/3 of all pregnancy-related deaths.
ZAHARA II Study type: Inclusion criteria: 1 endpoint: Cardiovascular events Postpartum risk is low among women
Kampman et al. Prospective cohort Pregnant women with within 1 y postpartum free of events during pregnancy
2015 (468) known congenital heart Women who have events during
25641540 Size: 172 disease Results: Women with events during pregnancy should be followed
Exclusion criteria: N/A pregnancy were 7.1 times more likely postpartum for changes in cardiovascular
to have events postpartum status.

259

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Arrhythmias were most common
events, mostly atrial; others not specified
ZAHARA Study type: Inclusion criteria: 1 endpoint: Cardiovascular events Presence of cyanotic heart disease
Drenthen et al. retrospective Pregnant women with during pregnancy (corrected/uncorrected), use of cardiac
2010 (469) analysis of registry known congenital heart medication before pregnancy, left heart
20584777 data disease Results: Cardiovascular complications obstruction, aortic or pulmonic
Exclusion criteria: occurred in 7.6% of pregnancies, with regurgitation, and mechanical valves
Size: 1302 Miscarriages at <20 wk clinically significant arrhythmias were most closely associated with
pregnancies in 714 of gestation; elective most common events 4.7%; type cardiovascular complications.
women with abortions. not specified.
congenital heart
disease
Mhyre et al. 2014 Study type: Inclusion criteria: 1 endpoint: Cardiac arrest during CPA is rare among patients hospitalized
(470) Retrospective Diagnosis code hospitalization for delivery in the for delivery, but considerably higher than
24694844 cohort study of CA indicating delivery or a United States between 1998 and the age adjusted incidence of CPA in
during admissions procedure code related 2011. 2 outcomes included: (1) general population.
for delivery from to delivery survival to hospital discharge; (2) the There is a trend towards improving
the Nationwide association between CA and survival to hospital discharge over the 14
Inpatient Sample Exclusion criteria: demographic and socioeconomic y observation period, but the incidence
(NIS) Diagnosis code characteristics, and medical and has not changed significantly.
indicating abnormal obstetric diagnoses and procedures; The most common etiologies
Size: 56,900,512 products of conception and (3) association between CA and numerically are those that are not
hospitalizations for or a procedure code the annual hospital delivery volume. associated with the tachyarrhythmic CA,
delivery between indicating abortion. but the incidence is highest among those
1998 and 2011 Results: 4,843 cardiopulmonary conditions that are more likely to be
arrests (CPA) between 1998 and 2011 associated with tachyarrhythmic events.
(event rate = 8.5 CPA/100,000 The cumulative number of CPAs in the
hospitalizations, or 1: 12,000). sample was 4,843 over 14 y (average =
Incidence was higher for older 346/y), but this number is based on the
subjects (>35 y), black women, and limitations of the sample size in the NIS.
Medicaid patients. The conditions
most strongly associated with CPA
were pulmonary hypertension,
malignancy, CVD (i.e., ischemic heart
disease, congenital heart disease,
cardiac valvular disease, and pre-
existing hypertension), liver disease,

260

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
and systemic lupus erythematosus.
However, the absolute numbers were
highest for postpartum or
antepartum hemorrhage combined =
44.7%, HF, amniotic fluid embolism,
and sepsis.
Siu et al. 2001 Study type: Inclusion criteria: 1 endpoint: Prepartum (2nd and 3rd A subgroup at high risk for 1 or 2
(471) Retrospective Congenital or acquired trimesters), peripartum, and cardiac complications of pregnancy is
11479246 analysis of a cardiac lesions or postpartum 1 cardiac, 2 cardiac, identifiable, with a combined incidence of
multicenter cardiac arrhythmias. neonatal, or obstetric complications. 17%. Among 1 events, 55% occurred
consecutive series Patients in whom during the 2nd and 3rd trimesters.
of pregnant women cardiac arrhythmia was Results: The majority of arrhythmias were SVTs.
with a Hx a heart the 1 diagnosis must The principal cardiac lesion was Careful scrutiny of high risk cardiac
disease. have had symptomatic congenital in 445 pregnancies (74%), patients during pregnancy, beginning no
sustained acquired in 127 pregnancies (22%), later than the second trimester, is
Size: tachyarrhythmias or and arrhythmic in 27 pregnancies warranted for both arrhythmic and non-
599 pregnancies in bradyarrhythmias (4%, with the majority being SVTs). arrhythmic 1 and 2 complications.
562 consecutive requiring treatment 1 cardiac events occurred in 80
referrals before pregnancy. pregnancies (13%); 55% of which
Exclusion criteria: occurred prepartum. Pulmonary
Isolated mitral valve edema and/or cardiac arrhythmia
prolapse (moderate or accounted for most of the cardiac
mild mitral events, the majority SVTs. Predictors
regurgitation) or those of 1 cardiac events were HF, TIA,
referred for CVA, or arrhythmia before
termination of pregnancy; baseline NYHA class >II or
pregnancy. cyanosis; left heart obstruction; and
LV EF<40%.
A 2 cardiac event occurred in 37
(6%). Worsening of NYHA class by >2
classes occurred in 26 of the 579
pregnancies in which the baseline
NYHA class was I or II.
Einav et al. 2012 Study type: Inclusion criteria: 1 endpoint: Maternal outcomes may not be as poor
(472) Retrospective (1) At least 5 clinical Maternal and neonatal survival to as in other CA populations.
22613275 analysis of details regarding the hospital discharge and the Mortality rates were higher among
published original case (e.g. age, women who underwent PMCD compared

261

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
articles, case series, gravidity, parity, relationship between PMCD and this with those who did not, possibly because
case reports and obstetric and medical outcome. of a subgroup with spontaneous or rapid
letters to the editor Hx, presenting rhythm, Results: ROSC.
regarding PMCD location of arrest), and ROSC was achieved in 60.6% of The 4-min time goal for PMCD usually
during CA in the care provided (e.g. mothers (N=57), among whom 89.5% remains unmet (4 of 57, 7%), yet neonatal
pregnancy chest compression, survived to hospital discharge survival is still likely if delivery occurs
Size: ventilation, monitoring, (51/57). Time from arrest to PMCD within 10 or even 15 min of arrest and
94 cases selected drugs given); (2) At was reported for only 57 cases of the neonatal survival was most-powerfully
from 108 least one of the 76 (75%) receiving PMCD; the associated with maternal arrest occurring
publications that following outcomes: (a) average time was 16.612.5 min in-hospital, regardless of the cause of
met review criteria. maternal non- (median 10, range 160, IQR 825), arrest.
return/return of with only 4 cases achieving the
spontaneous recommended 4-min target.
circulation or non- Overall survival to hospital discharge
survival/survival to was 54.3%. Among 23 with VT/VF, 15
hospital discharge; (b) survived to discharge. Overall, in-
fetal/neonatal hospital location and PMCD <10 min
outcome. were statistically significant.
Neurological outcomes of surviving
Exclusion criteria mothers (N=51) were described as
Maternal arrest post- CPC 1/2 in 78.4% (40/51).
delivery, no data The overall neonatal survival rate was
enabling relation of 63.6% (42/66). Neurological
case details to outcomes of surviving neonates were
outcome, or if both CPC 1/2 in 52.3% (22/42),
outcomes were
unclear.
Citro et al. 2013 Study type: Case Inclusion criteria: 1 endpoint: Diagnosis of TTS Acute medical/surgical stressors are
(473) reports identified in Diagnostic criteria for increasingly recognized as a trigger for
23519095 systematic tako-tsubo syndrome Results: 13 of 15 cases of TTS had TTS
literature review based upon modified onset 24 h after a C-section. Distinction from peripartum
Mayo criteria cardiomyopathy is important for
Size: 15 13 patients had cardiac complications prognostic reasons.
Exclusion criteria: (pulmonary edema, cardiogenic Cardiac arrest is infrequent in TTS.
Preexisting shock, or CA [N=1]) LQT2 more likely to have ACE
cardiomyopathy or All patients had return of LV function postpartum vs LQT1 or 3
in 13.4310.96 d.

262

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
other known cardiac Risk greatest during 9 mo postpartum:
defects HR: 2.7, 95% CI: 1.84.3, p<0.001
risk reduced by using beta-bl, HR: 0.34,
95% CI: 0.14-0.84, p=0.02.
Seth et al. 2007 Study type: Inclusion criteria: 1 endpoint: LQTS-related death, The data have implications for
(474) Retrospective First live birth ACA, and/or syncope before, during, observation and pharmacological
17349890 analysis of data pregnancy in women and after pregnancy management during the 9 mo post-
from the with identified LQTS- partum.
International LQTS related gene mutation Results:
Registry or considered to be Compared to frequency of endpoint
affected with LQTS on events prior to pregnancy, event
Size: the basis of a QTc>470 rates during pregnancy were lower,
391 ms but significantly higher during the 9
Exclusion criteria: mo postpartum period. Frequency of
First live birth prior to events returned to pre-pregnancy
1980. levels after 9 mo. The post-partum
increase was greatest among those
with HERG mutations.
Katz et al. 2005 Study type: Inclusion criteria: 1 endpoint: The data reviewed supports, but does
(475) Systematic Case reports of Outcomes for fetus and mothers as a not prove, that PMCD within 4 minutes of
15970850 MEDLINE review of pregnant CA victims result of PMCD onset of maternal CA improves maternal
outcomes from between 25 and 42 wk and neonatal outcomes.
perimortem of gestation who Results: A controlled trial will never be feasible.
cesarian deliveries underwent PMCD. In 30 of 38 PMCDs surviving infants The conclusion is based upon general
Size: were delivered. One of the twins died data on survival free of neurological
38 Exclusion criteria: in the neonatal period from anoxic injury during CA as a function of down-
Cesarean deliveries injury and complications of time.
performed on mothers prematurity. In 12 of 22 cases in
who were dying from which hemodynamic data was
mortal injuries, but still reported, sudden return of pulse and
had vital signs, were BP occurred when the uterus was
excluded. emptied.
Dijkman et al. Study type: Inclusion criteria: 1 endpoint: Use of PMCD is increasing over time.
2010 (476) Retrospective All cases of maternal Frequency of use of PMCD over time Outcome for pregnant women with CA
20078586 cohort study of CA CA during the second and case fatality rate of those with and PMCD remains dismal, but this study
during pregnancy, half of pregnancy in PMCD (N=12) compared to those is limited by small numbers and apparent
with and without The Netherlands without PMCD (N=43). long delays to initiation of PMCD.

263

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
PMCD during a 15 y identified by survey The data are reasonable for trend to
period. from 1993-2008. Results: increased used of PMCD, but outcomes
A total of 8 of 55 mothers survived cannot be relied upon because of factors
Size: Exclusion criteria: (15%). Among the 12 women in cited above.
55 CA among None specified whom PMCS was performed, there
2,929,289 women, were two maternal survivors (17%).
12 of whom In the 43 women in whom no PMCS
underwent PMCD. was performed, there were six
maternal survivors (14%).
No PMCDs were performed prior to
2000, and the use progressively
increased after 2000. The maternal
case fatality rate for PMCS for the
entire 15 y period was 83% (10/12).
For the period of August 2004 to
August 2006 the case fatality rate for
PMCS was 75% (3/4) and the case
fatality rate for resuscitation without
PMCS was 67% (6/9).
Neonatal case fatality rate with
PMCD was 58%. Corresponding data
for no PMCD is not provided.
Colletti et al. 2013 Study type: Inclusion criteria: 1 endpoint: Even in light of these numbers, it is
(477) Review and opinion Studies of radiation Magnitude of exposure risk to fetus generally recommended that fluoroscopic
23436839 article on radiation exposure to fetus as a based upon nature of radiation- procedures be avoided until after the first
during pregnancy result of cardiovascular associated procedure and stage of trimester, unless clinical circumstances,
procedures in pregnant pregnancy based on risk/potential benefit
Size: women. considerations, warrant an earlier
Not specified Results: intervention.
Exclusion criteria: Most procedures entail a fetal dose
N/A well below the fetal risk threshold of
50 mGy. For the specific issue of
fluoroscopic radiation for ICD
implants, no specific data is available.
However, for groin-to-heart catheter
procedures, the fetal exposure is
0.0940.244 mGy/min. Thus, a

264

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
fluoroscopic time of 1 h falls well-
below the fetal risk threshold.
Natale et al. 1997 Study type: Inclusion criteria: 1 endpoint: ICDs are effective and safe for the
(478) Multicenter Women with an ICD Use, efficacy and safety of ICDs pregnant female
9386142 retrospective who completed a during pregnancy. There were no apparent adverse effects
analysis of women pregnancy or was Results: on the fetus.
with an ICD who currently pregnant. The EF at the time of ICD
became pregnant. (1). The clinical implantation was 49.89.7% (present
presentation and EF was 51.49.5%). Underlying
Size: indication for ICD cardiac diseases were long-QT
44 implantation were syndrome (N=13), idiopathic VF (17),
sudden cardiac death cardiomyopathy (8), congenital heart
in 33 patients, VT in 9 disease (3), CAD with an ischemic
patients, and VT with cardiomyopathy (1), HCM (1), and
syncope in 2 patients. ARVC (1). The indications for the ICD
were VF in 33 patients, VT in 9, and
Exclusion criteria: N/A VT/syncope in 2.
During the first pregnancy after
implant, 33 women experienced no
ICD discharge, 8 received one shock;
1 experienced 5 firings in Afib; and 2
had 11 and 5 discharges, respectively,
for monomorphic VT. During delivery,
in the women in whom the ICD
remained active, none received any
shocks. In the 24 to 48 h period after
delivery, 1 patient had an ICD
discharge for VF. Overall, the total
number of ICD discharges during
pregnancy ranged from none to 11,
with an average of 0.661.9 shocks
(0.07 shock per mo).
There were no apparent adverse
effects on the fetus among the 11
shocks delivered during pregnancy

265

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Damilakis et al. Study type: Inclusion criteria: 1 endpoint: Catheter ablation procedures result in a
2001 (479) Radiation exposure Women of childbearing Radiation exposure and fluoroscopy very small increase in risk of potentially
11514375 and fluoroscopy age undergoing times estimated for phantom harmful radiation effects to the fetus.
tines to a catheter ablation simulated fetus, calculated for first,
theoretical fetus procedures for second, and third trimesters.
during simulated supraventricular Results:
pregnancies during tachycardias. The average radiation dose to the
ablation procedures fetus was <1 mGy in all periods of
in female patients Exclusion criteria: gestation. Average excess fatal cancer
of childbearing age. N/A was 14.5/106 fetuses exposed during
Estimated radiation the first trimester. Corresponding
exposure was values for the second and third
carried out for each trimesters were 30 and 55.7/106,
projection of the respectively. The risk for hereditary
cardiac ablation effects in future generations was
procedure, using 1.5/106 cases for irradiation during
fetal phantoms the first trimester. Corresponding
simulating values for the second and third
pregnancy in the trimesters were 3.0 and 5.6/106,
first, second, and respectively.
third
trimesters.

Size: 20 women

266

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 52. RCTs Comparing Medication-Induced Arrhythmias - (Section 10.7)
Study Acronym; Aim of Study; Patient Population Study Intervention Endpoint Results Relevant 2 Endpoint (if
Author; Study Type; (# patients) / (Absolute Event Rates, any);
Year Published Study Size (N) Study Comparator P values; OR or RR; & Study Limitations;
(# patients) 95% CI) Adverse Events
CAST Aim: Test Inclusion criteria: Intervention: Drugs as 1 endpoint: after 10 Excess in deaths due to
The Cardiac hypothesis that Post MI, 6 d to 2 y; six listed mo there was an excess shock due to recurrent MI.
Arrhythmia suppression of or more PVCs/h and Encainide 432, placebo in deaths due to
Suppression Trial ventricular ectopy no VT over 15 beats at 425 arrhythmia (p=0.0004)
Investigators. 1989 post MI reduces 120 bpm. 80% Flecainide 323, placebo in patients treated with
(480) incidence of SCA n suppressioin of PVCs 318. encainide or flecainide.
2473403 patients whose and 90% suppression
ectopy was of NSVT. Comparator: Placebo
suppressed by Safety endpoint (if
encainide, Exclusion criteria: No relevant): n/a
flecainide or flecainide for EF<30%.
moricizine Moricizine was second
choice if EF>30%
Study type:
Randomized
contolled, double-
bllind

Size: 1498
CAST II Aim: test Inclusion criteria: Intervention: Moricizine 1 endpoint: N/A
The Cardiac hypothesis that Post MI, 6 d to 2 y; six Terminated early due
Arrhythmia suppression of or more PVCs/h and Comparator: Placebo, to excess mortality (17
Suppression Trial II ventricular ectopy no VT over 15 beats at of 665 with death or
Investigators. 1992 post MI reduces 120 bpm. 80% SCA with moricizine vs
(481) incidence of SCA n suppressioin of PVCs 3 of 660 with placebo)
1377359 patients whose and 90% suppression
ectopy was of NSVT. Safety endpoint: n/a
suppressed by
moricizine Exclusion criteria:
patients with any runs
lasting 30 sec or

267

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Study type: longer at a rate of
Randomized 120 complexes/min
contolled, double-
bllind

Size: 1335

Data Supplement 53. Nonrandomized Trials, Observational Studies, and/or Registries of Medication-Induced Arrhythmias (Section 10.7)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Wyse et al. 2001 Study type: Inclusion criteria: 1 endpoint: Mortality Mortality of patients with a transient
(482) Prospective study of Patients with transient or correctable cause of VT/VF was no
11704386 the registry of AVID, or correctable VT/VF, Results: mortality of patients different or perhaps even worse than
examining the compared with patients with a transient or that of the 1 VT/VF.
outcome of patients with high risk in AVID correctable However, the small number of patients
with transient or registry. Patients in cause of VT/VF was no with AAD reaction seemed to most
correctable causes registry could have EF different or perhaps even likely to presage better survival
of VT/VF >40% worse than that of the 1
VT/VF.
Size 278 patients with Exclusion criteria: N/A
transient or
correctable cause, of
4450 in registry; only
18 (6.5%) had an AAD
reaction
Monnig et al. 2012 Study type: Single Inclusion criteria: 1 endpoint: ICD shock ICD therapy was appropriate in 44% of
(483) center observational survival of CA due to patients with drug-induced QT
21979994 trial acquired QT Results: Over mean followup prolongation/TdP, (where DI-TdP was
prolongation/TdP who of 84 mo, 44% had due to an AAD in 79%).
received an ICD. 79% had appropriate shocks and
drug-induced TdP from inappropriate shocks in 30% However, EF was not normal (mean
an AAD. sotalol N=17; (Only inappropriate in 3 of 43) 4112)
Size 43 patients amiodarone N=12;
quinidine

268

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
N=3; propafenone N=1; Appropriate shocks were most
ajmaline N=1] common in those with structural disease.

Exclusion criteria: N/A Beta blockers did not seem to reduce


risk
Antman et al. 1990 Study type: An open- Inclusion criteria: 1 endpoint: Resolution of 90% of patients had a treatment
(484) label multicenter Digitalis intoxication with toxicity and time course. response in the setting of advanced and
2188752 clinical trial of Fab actual or potentially Dosing requirements potentially life-threatening digitalis
treatment for life- life-threatening cardiac toxicity.
threatening digitalis rhythm disturbances, Results: 80% had resolution
intoxication hyperkalemia, or both of all signs and symptoms of
caused by digitalis toxicity, 10% improved, and
intoxication; refractory to 10% showed no response.
or likely to be refractory Median initial response time
to treatment with was 19 min. Time to complete
Size 150 conventional therapeutic response was 88 min median
modalities. (30360 min).
46% had refractory VT 54% of those with CA survived
and 33% had VF. hospitalization.
Adverse events in 14/148,
Exclusion criteria: N/A with hypokalemia or
worsening CHF.
Chan et al. 2014 Study type: Review Inclusion criteria: digoxin 1 endpoint: Resolution of Confirms efficacy, onset of action.
(485) of 10 case series poisoning toxicity, time course to effect. Suggests that lower doses (at lower cost)
25089630 are appropriate in many situations due to
Exclusion criteria: N/A Results: Response varied pharmacokinetics of digoxin (unless CA is
from 80-90% to 50%. imminent).
Reversal of toxicity 3045
Size 2080 min.
Adverse events <10%
(exacerbated CHF, increased
HR and hypokalemia)
Lower dose requirements
(1/2 of the full neutralizing
dose) are appropriate unless
CA is imminent.

269

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Hauptman et al. 1999 Study type: Review Inclusion criteria: N/A 1 endpoint: N/A More common manifestations
(486) of treatment of (including occasional ectopic beats,
10069797 digoxin toxicity Exclusion criteria: N/A Results: N/A marked first-degree AV block, or AF with
a slow ventricular response) require only
temporary withdrawal of the drug and
Size N/A monitoring.

Administration of potassium salts is


recommended for ectopic VA, even
when the serum
potassium is within the normal range.
Kelly et al. 1992 (487) Study type: Review Inclusion criteria: N/A 1 endpoint N/A Describes VT with digoxin toxicity.
1626485 Notes exacerbation of digoxin toxicity
Exclusion criteria: N/A Results: N/A with low and high K, hypothyroidism,
Size: N/A Notes benefit of magnesium
administration.
Osmonov et al. 2012 Study type: Single Inclusion criteria: drug- 1 endpoint: improvement or Digoxin-induced AV block (without
(488) center observational related symptomatic type need for pacer. toxicity) usually improved (28 of 39)
22530749 series. 2 second degree or third after withdrawal of the drug.
degree AV block Results: 39 patients had AV
block with digoxin dosing,
Exclusion criteria: MI, with 28 of them improving
electrolyte abnormalities, after withdrawal of the drug.
Size: 108 digitalis toxicity, and
vasovagal syncope.
Digoxin toxicity (a digoxin
level from a blood
test of higher than 2
nmol/L with symptoms
such as nausea,
vomiting, and color vision
abnormalities or
Above 2.5 nmol/L with or
without symptoms.
Tzivoni et al. 1988 Study type: Inclusion criteria: TdP 1 endpoint Abolition of TdP This established MgSO4 as treatment
(489) Consecutive series (9/12 due to AAD) for TdP

270

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
3338130 Provided 2 gm IV with Results: In nine of the
second bolus of 2 g Exclusion criteria: N/A patients a single bolus of 2 g
after 5-15 min. 9 completely abolished the TdP
received infusion at 3- within 1 to 5 min, and in three
20 mg/min for 7-48 h. others complete abolition of
the TdP was achieved after a
Size 12 second bolus was given 5 to
15 min later.
Keren et al. 1981 Study type: Single Inclusion criteria: TdP, 1 endpoint: response to This confirmed the effectiveness of V
(490) center series QTc>600 ms therapy of isoproterenol pacing for DI-TdP, even after
7296791 and/or ventricular pacing. isoproterenol was ineffective.
Exclusion criteria: N/A
Results: Pacing effective in 4 This confirms the effectiveness of
of 4 patients, 2 who had not isoproterenol as a first line treatment.
Size: 10 (9 on AAD, 4 responded to isoproterenol.
treated with pacing) Continued up to 48 h and Magnesium was not given in this
pacer removed after another series.
24 h. Pacing rate was lowest
effective rate, 88-105 bpm.

In 2 cases atrial pacing was


tried, initially effective but
unstable so V pacing
provided.

Lidocaine was given in 4 cases


without improvement.

Isoproterenol (2-8
microgram/min) was given in
7 cases: effective in 5/7.
Choy et al. 1997 Study type: Inclusion criteria: healthy 1 endpoint: Effect on QTUc Potentially arrhythmogenic QT
(373) Double-blind subjects (12) and CHF from KCl after quinidine or abnormalities during quinidine treatment
9337183 comparison of (mean EF 17%) with age- placebo. and in CHF can be nearly normalized by
potassium infusion matched controls modest elevation of serum potassium
after quinidine and without CHF Results:

271

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
placebo sequentially in Exclusion criteria: N/A KCl was IV, 0.5 mEq/kg (to
12 healthy subjects. maximum of 40 meEq) over
Also, study on QTU in 60-70 min resulted in
patients with CHF and normalization of quinidine-
age-matched controls induced and CHF-related QTU
who receive IV KCl prolongation

Size: 12 healthy, 8
CHF plus 8 age-
matched controls
Yang et al. 1996 (491) Study type: Basis EP Inclusion criteria: N/A 1 endpoint: Change in IC50 Extracellular potassium is a critical
8565156 (cardiac myocytes) for dofetilide and quinidine determinant of drug block of IKr, with
Exclusion criteria: N/A according to the extracellular substantial clinical implications. The
K concentration increase in drug block with low [K+]o
provides a mechanism to explain the link
Results: Elevating [K+]o from between hypokalemia and torsade de
Size: N/A 1 to 8 mmol/L increased the pointes
IC50 for dofetilide block from
2.70.9 to 7932 nmol/L and
for quinidine block from
0.40.1 to 3.81.2
mol/L.Increased K blunted
drug effect of dofetilide and
quinidine
Hellestrand et al. Study type: Clinical Inclusion criteria: Group 1 endpoint: Flecainide significantly increased both
1983 (492) research study I:11 with temporary acute and chronic thresholds and the
6195608 pacer; Group II:10 with Results: Given IV flecainide 2 most marked rise (>200%) occurred
chronic pacer at mg/kg over 10 min. 7 with during chronic oral therapy.
generator change; Group programmable pacers given
III: 7 with programmable oral 100-400 mg per day.
Size: 28 pacer with pacing I: 0.661.44 V
threshold testing II: 1.732.13 V
III: 10 min: at 2.7 V: 0.140.22
Exclusion criteria: N/A msec; at 4.9 V 0.060.11
After 3 wk: at 2.7V 0.090.28
msec, at 4.9 V 0.060.16

272

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Echt et al. 1989 (493) Study type: Basic Inclusion criteria: N/A 1 endpoint: change in Lidocaine doubled the defibrillation
2469545 canine study defibrillation threshold (DFT) energy requirement
Exclusion criteria: N/A
Size: 78 protocols Results: ED90 increased from
total 11 to 22 Joules (p<0.01)

Crijns et al. 1988 Study type: Inclusion criteria: Rate 1 endpoint: N/A Wide complex tachycardia resulted
(494) observational trial related BBB giving wide from tachycardia and flecainide slowing
3143257 QRS tachycardia Results: 6 patients developed conduction. This can appear to be VT but
Size: 6 of 79 patients WCT, rates 145-200 BPM is not.
treated with flecainide Exclusion criteria: N/A
developed this wide
complex tachycardia
Bajaj et al. 1989 (495) Study type: Basic Inclusion criteria: N/A 1 endpoint: After infusion of Short-term administration of NaHCO3
2551538 canine ODE, a potent metabolite of or NaCl can partially reverse ODE-
Exclusion criteria: N/A encainide, shortening in induced conduction
intervals (HV and QRS) with slowing, which may be an important
NaHCO3 or NaCl factor in arrhythmia aggravation

Size: 30 Results: With NaHCO3, QRS:


9276 msec; HV 44 to 37
msec.
Myerburg et al. 1989 Study type: Case Inclusion criteria: Prior 1 endpoint: suppression of Propranolol had failed to prevent
(496) series CA or symptomatic drug-induced arrhythmias inducibility of sustained VT during
2480856 sustained VT, treated previous programmed stimulation
with a Ic medication who Results: Drug-induced studies in three of the four patients, but
developed runs of arrhythmias were suppressed it reproducibly
sustained VT, NSVT or in all 4 patients suppressed drug-induced arrhythmias
Size: 4 (3 flecainide, 1 increased ectopy that appeared only after administration
encainide) of the IC agents in each patient.
Exclusion criteria: N/A
Schwartz PJ et al. Study type: Review Inclusion criteria: N/A N/A Review of Hx of drug-induced QT
2016 (497) prolongation and TdP.
27150690 Exclusion criteria: N/A crediblemeds.org categorizes drugs as
possible, conditional and known TdP risk.
Drugs associated with prolonged QT
and TdP fall into a number of different
273

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
pharmacologic classes, and the risk of
TdP increases according to clinical and
genetic factors.
Clinical decision support systems
reduce prescription of QT prolonging
drugs in patients at risk of TdP due to
clinical or genetic factors.
Kannankeril P, et al. Study type: Review Inclusion criteria: N/A 1 endpoint N/A Hypokalemia worsens risk of TdP
Pharcological Reviews Although no randomized prospective trial
2010. (374) Size: N/A Exclusion criteria: N/A Results: N/A has been conducted, intravenous
magnesium has become a first-line
therapy for drug-induced TdP.

Data Supplement 54. Nonrandomized Trials, Observational Studies, and/or Registries Related to ACHD - (Section 10.8)
Study Acronym; Study 1 Endpoint and Results
Summary/Conclusion
Author; Type/Design; Patient Population (P values; OR or RR;
Comment(s)
Year Published Study Size & 95% CI)
Basso C, et al. Study type: Inclusion criteria: N/A 1 endpoint: N/A Discussed gross and microscopic
Virchows Arch Review Role of autopsy to establish cause of SCD: Assoc of pathologic findings
2008 (498) Exclusion criteria: N/A European Cardiovascular Pathology developed
17952460 Size: N/A guidelines Further tests in future:
Includes ARVC, athletes heart, HCM, myocarditis molecular or toxicology

Results: N/A
Thorne SA, et Study type: Inclusion criteria: 1 endpoint: Review side effects of chronic oral Patients with CHD at higher risk
al. Retrospective ACHD, mean age 34.9 amiodarone for amio adverse effects, esp
Circ 1999 (499) multicenter y, receiving women, cyanosis, Fontan, or dose
10402444 amiodarone for 6 mo; Results: 36% developed thyroid dysfunction: 19 >200 mg
Size: 92 pts case-control group. hyper, 14 hypothyroid. Sig risk factors: Female
Mean duration 3 y, gender (OR: 3.0) cyanotic HD (OR: 7.0); Fontan
mean dose 191 mg (OR: 4.0); dosage >200 mg/d (OR: 4.0)
Exclusion criteria: N/A
Deal B, et al. Study type: Inclusion criteria: TOF 1 endpoint: Induction of VT in TOF, response to TOF EPS reproduces clinical
AJC 1987 (500) single center pts undergoing cath + drug rx sustained VT
3591695 retrospective EPS and drug testing Mean 3.3 drugs/pt tested. Followup mean 2.2 y Pts with freq PVCs: 60% inducible
Sust VT: 4 sust VT
274

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 9 PVCs: 5 Results: all pts with clinical sust VT had inducible Surgery to improve hemodynamics
sustained VT eliminated VT
Exclusion criteria: 60% pts with frequent PVCs had inducible sust VT Elevated RV pressure: did not
Pts with RV hypertension did not respond to any respond to medicationss
medications
4 pts underwent surgery: no recurrent VT
Gatzoulis MA et Study type: Inclusion criteria: TOF 1 endpoint: TOF mechano-electrical interaction TOF: QRS duration 180 msec
al. Circ 1995 Single center survivors Mean followup 24 y predicts VT and SCD
(501) prospective All patients with documented
7600655 Exclusion criteria: N/A Results: 41/178 patients evaluated serially, + sustained VT and patients with SCD
Size: 41 reviewed 4 SCD had QRS duration 180 msec
QRS duration correlated with RV size on Echo and (100% sensitivity)
heart size on CXR Chronic RV volume overload
VT 9 patients: QRS mean 199 msec, CTR 0.67; related to diastolic dysfunction
significantly different than those without VT
Koyak Z et al. Study type: Inclusion criteria: 1 endpoint: SCD in ACHD Risk for SCD in ACHD: SVT (OR:
Circ 2012 (502) Retrospective ACHD patients in 3.5), mod-severe systemic
22991410 multi-center Canadian database Results: 1,189 deaths among 25,790 ACHD ventricular dysfunction (OR: 3.4),
with case- patients: mod-severe sub-pulmonary vent
controls Exclusion criteria: N/A 19% SCD (213 patients) dysfunction (OR: 3.4), increased
Arrhythmic cause 80% QRS duration (OR: 1.34 per 10 msec
Size: 213 SCD vs severity of congenital heart disease increase)
Mild 12%, mod 33%, severe 55%
Diller GP et al. Study type: Inclusion criteria: TOF 1 endpoint: TOF: sustained VT, ACA/SCD, approp TOF: sust VT/SCD1.2/ACA 4.6%
Circ 2012 (503) Single center patients ICD shock LV longitudinal function
22496160 retrospective Mean age 36 y associated with greater risk SCD/VT
Median followup 2.9 y Results: 4.6% sust VT/SCD/ACA
Size: 413 (SCD 1.2%, Sustained VT, 2.2%, ICD shock 1.2%)
Exclusion criteria: N/A Combination echo variables c/w poor outcome: RA
area, RV fractional area change, LV global
longitudinal strain, mitral annular systolic
excursion
Harrison DA et Study type: Inclusion criteria TOF 1 endpoint: TOF and sustained VT TOF patients with VT have
al. JACC 1997 Single center and VT, compared anatomic aneurysms of RVOT or PR
(504) retrospective with 192 TOF patients Results: Patients with VT had frequent PVCs, low Combined approach of correcting
9350941 without arrhythmia CI, RVOT aneurysms/PR/TR structural abnormalities + intra-op
Size: 18 map-guided VT ablation may
275

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: N/A 14 patients reoperated: 10/14 cryoablation map- reduce risk of deteriorating
guided: recurrent VT in 3/10 function and optimize VT
Two patients with VT developed severe CHF, died. management

Knauth Al et al. Study type: Inclusion criteria: TOF 1 endpoint: TOF major ACE: death, sustained VT, TOF adverse outcomes predictors:
Heart 2008 (505) Single center patients with CMR NYHA Class III/IV, clinical predictors RVEDV z score 7, OR: 4.55
17135219 retrospective Results: MACE: 20.5%: death 5%, Sustained VT LVEF <55%, OR: 8.05
Median postop 10%, worsening NYHA class 11% RVEF <45%
Size: 88 interval: 21 y QRS duration 180 msec correlated with RV size QRS duration 180 msec
Exclusion criteria: N/A
Therrien J et al. Study type: Inclusion criteria: PVR 1 endpoint: Impact of PVR in TOF on QRS PVR in TOF:
Circ 2001 (506) cohort study for TOF duration and VT, AT QRS duration stabilized
11369690 Mean followup 4.7 y Concurrent cryoablation decreased
Size: 70 VT preop 22% Results: Cryoablation 15 patients with intraop incidence of VT
AT preop 17% mapping: 9 VT, 6 AFL: none had recurrence of pre-
existing arrhythmia
Exclusion criteria: N/A VT post PVR 9% from 22%, p<0.001
AFL/AF decreased from 17% to 12%, p=0.32
Therrien J et al. Study type: Inclusion criteria 1 endpoint: TOF and PVR: effect on RV volume TOF and PVR:
AJC 2005 (507) Single center adult TOF undergoing Mean followup 21 mo Decreases RV volumes
15757612 retrospective pulmonary valve Results: PVR decreased RV volume: RVEF did not change
replacement (PVR) RVEDV: From 163 ml/m2107 ml/m2 PVR before marked RV volume
Size: 17 RVESV: 109 to 69 ml/m2 increase?
Exclusion criteria: N/A RVEF did not change: EF 3234

Patients with RVEDV >170 ml/m2 or RVESV >85


ml/m2: no pt had normalization of RV volume after
surgery
Harrild DM et Study type: Inclusion criteria TOF 1 endpoint: Impact of PVR in TOF on major TOF with late PVR: VT or death
al. Circ 2009 Single center patients with late adverse events every 20 patient-y
(508) retrospective pulmonary valve followup median 1.4 y In matched comparison with TOF
19139389 replacement for RV controls, PVR did not reduce the
Size: 98 dilation; matched Results: Freedom from death or VT: incidence of VT or death
controls with TOF, RV 5 y: 80%, 10 y: 41% NOTE: advanced RV enlargement,
dilation but no PVR empiric cryoablation

276

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Median age 21 y Empiric cryoablation: 7 patients: 5/7 VT during
6% preop VT followup
QRS duration >180 Incidence death, VT, or both: 4.8/100 pt yrs
msec: 19% All cause mortality: 6.1%
Exclusion criteria: N/A No sig change in QRS duration after surgery
Adamson L et Study type: Inclusion criteria PVR 1 endpoint: Effect of PVR in TOF on RV size and PVR in TOF:
al. Interact CTS meta-analysis after TOF repair: 19 function Low mortality
2009 (509) medline 1950- papers analyzed Reduces RV volumes
19567499 2009 Results: summarizes all 19 papers conclusions RV function improves
Exclusion criteria: N/A Symptoms and functional status
Size: 1070 improves
Sabate Rotes A Study type: Inclusion criteria: TOF 1 endpoint: Impact of PVR in TOF on major TOF and PVR:
et al. CAE 2015 Single center patients with late adverse events: VT, SCD/ACA, appropriate ICD Hx of VT and LV dysfunction
(510) retrospective pulmonary valve shock associated with higher risk, HR: 4.7
25416756 replacement for RV QRS duration 180 msec
Size: 205 dilation between Results: Freedom from MACE: predictive of arrhythmic event
1988-2010 5 y: 95%, 10 y: 90%, 15 y: 79% Surgical cryoablation of VT may
Median age 33 y More events occurred in patients without be protective
Prior VT 8% cryoablation
LVEF <50%: 16% Cryoablation of VT: 22 patients: (11%) 1/22 event Recommend patients with risk
after 7 y. Empiric Cryo performed in patients with factors for VT undergo pre-or
Exclusion criteria: N/A VT, inducible VT at EPS not ablated, or Hx of postop EPS
unexplained syncope/pre-syncope; not map-
guided

Tsai SF et al. Study type: Inclusion criteria: 1 endpoint: Inducible VT in ACHD patients Inducible VT: 29%
AJC 2010 (511) single center ACHD patients 18y without clinical VA Combined fibrosis on MR and
20723654 retrospective undergoing V stim peak oxygen uptake <80%
Mean age 30 y Results: Inducible sust VT: 29% predicted had 100% sensitivity for
Size: 80 (TOF 52%, TGA 26%) sustained VT
Exclusion criteria: Predictors: increased QRS, decreased VO2 on Consider using MRI, ex test as
patients with clinical exercise, ventricular fibrosis on MRI (p < .05) screening for V stim studies
ventricular
arrhythmias

277

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Garson A et al. Study type: Inclusion criteria: TOF 1 endpoint: Induction of VT in TOF TOF with inducible VT: more
JACC 1983 (512) single center patients undergoing EP frequent PVCs, longer HV interval,
6853902 retrospective Results: patients with syncope had inducible elevated RV pressure, reduced RV
Exclusion criteria: N/A sustained or non-sust VT EF
Size: 27 Poor hemodynamics correlated
with VT induction
Chandar JS et Study type: Inclusion criteria: TOF 1 endpoint: Inducible VT in TOF Correlation poor hemodynamics
al. AJC 1990 Multicenter patients undergoing with inducible VT
(513) retrospective EPS Results: Induced VT correlated with delayed age
1689935 Mean age repair 5 y at repair, longer followup, syncope, elevated RV
Size: 359 Mean followup 7 y pressure, frequent PVCs on holter
Exclusion criteria: N/A
Koyak Z et al. Study type: Inclusion criteria: 1 endpoint: SCD in ACHD Risk for SCD in ACHD:
Circ 2012 (502) Retrospective ACHD patients in SVT (OR: 3.5)
22991410 multi-center Canadian database Results: 1189 deaths among 25790 ACHD mod-severe systemic ventricular
with case- patients: dysfunction (OR: 3.4)
controls Exclusion criteria: N/A 19% SCD (213 patients) mod-severe sub-pulmonary vent
Arrhythmic cause 80% dysfunction (OR: 3.4)
Size: 213 SCD vs severity of congenital heart disease increased QRS duration (OR: 1.34
Mild: 12%, mod: 33%, severe: 55% per 10 msec increase)

Kella DK et al. Study type: Inclusion criteria: ICD 1 endpoint: ICD outcomes in ACHD Non-TOF patients less likely to
PCE 2014 (514) Retrospective in ACHD patients Median followup 3.2 y receive appropriate shocks
24889130 single center TOF 56% ICD implantation indications
TGA 25% Results: 1 prevention 53% should be ACHD lesion specific
Size: 59 Approp ICD therapies 20%
Exclusion criteria: N/A 22% inapprop shocks
TOF: 27% approp shocks, non-TOF: 11% (p=0.043)
Santharam S et Study type: Inclusion criteria: 1 endpoint: ICD outcomes in ACHD ACHD and ICD:
al. Europace Retrospective ACHD patients with Mean followup 5 y 2.9%/y shock rate
2016 (515) single center ICD 2000-2014 Complications 9%/y
27234868 Mean age 41 y Results: Indications: Disease specific indications, risks
Size: 42 TOF 50%, TGA 12% 2 prev: 62% must be clearly discussed
1 38%. alternatives for 1 prevention
Exclusion criteria: N/A Appropriate shocks 14% ablation
Complications: 45%

278

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Vehmeijer JT et Study type: Inclusion criteria: 24 1 endpoint: ICD implants in ACHD High rate appropriate ICD therapy
al. EHJ 2016 Meta-analysis studies with 2162 Mean followup 3.6 y in both 1 and 2 ACHD
(516) EMBASE, ACHD patients with High rates inappropriate shocks
26873095 MEDLINE, ICD: Results: 1 53%, 2 47% and complications
Google Scholar Mean age 36 y Approp intervention (ATP or shock): 24%; Case-by-case analysis
TOF 50% 1 22%, 2 35%. costs/benefits essential
Size: 2162 Inapprop shocks 25%; Complications: 26%
Exclusion criteria: N/A All-cause mortality 10%
Moore JP et al. Study type: Inclusion criteria: 1 endpoint: Subcutaneous ICD in ACHD Subcut ICD feasible in ACHD, most
CAE 2016 (517) Retrospective subcut ICD in ACHD outcomes. Single ventricle 52%. commonly single ventricle patients
27635073 multi-center 7 starting 2011. Median followup 14 mo. with limited venous access
centers Median age 33.9 y Results: 1ary prevention: 67%, 2ary 33%. Successful conversion of induced
Implant: VT induced 81%, converted 80 joules in VT
Size: 21 Indication: limited all. Infection: 1 (5%); reasonable rhythm
venous access (10), Shocks: inapprop 21%, appropriate 1 (5%). One discrimination
right-to-left cardiac death due to asystole.
shunt 5
Exclusion criteria: N/A
Okamura H et Study type: Inclusion criteria: 1 endpoint: screening for suitability for for use of subcutaneous ICD in
al. Circ J 2016 Retrospective ACHD patients subcutaneous ICD use in ACHD patients ACHD, screening of left and right
(518) single center undergong screening Results: Left parasternal: failure 21%, reduced to parasternal position may improve;
27109124 for subcutaneous ICD 12% using right parasternal. QT interval and T wave inversion
Size: 100 Mean age 48 y V2-V6 independent predictors of
Exclusion criteria: N/A left parasternal screening.
Yap SC et al. Study type: Inclusion criteria: 1 endpoint: ICD outcomes in ACHD patients: ACHD Appropriate shocks 6%/yr,
EHJ 2007 (519) Multicenter ACHD patients 18 y median followup 3.7 y no difference in 1 or 2 prevention
17030523 retrospective, receiving ICD Inappropriate shocks 41%
Dutch national Mean age 3713 y Results: Early comps 13%, late 17%
registry 2 prevention 60% Approp shocks 23%, inapprop 41% -mainly SVT.
TOF fewer approp shocks vs other congenital heart
Size: 64 Exclusion criteria: disease, HR 0.29
Khairy P et al. Study type: Inclusion criteria: TOF 1 endpoint: TOF: correlate V stim with outcomes Multivariate analysis: inducible
Circ 2004 (520) Multicenter patients undergoing V Results: sust monomorphic VT 30%, polymorphic sustained VT independent risk for
15051640 cohort stim VT 4.4% subsequent clinical VT or SCD (RR:
followup 6.5 y Independent risk factors: age 18 y (OR: 3.3), 4.7)
Size: 252 palpitations (OR: 2.8), frequent PVCs (OR: 5.6), CT
Exclusion criteria: N/A ratio 0.6, prior shunt (OR: 3.1)

279

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Older age, prior shunts, frequent
PVCs, cardiomegalyincreased
likelihood of inducible VT
Khairy P et al. Study type: Inclusion criteria: TOF 1 endpoint: TOF ICD outcomes TOF ICD shocks annual rate 7.7
Circ 2008 (521) Retrospective patients receiving ICD Median followup 3.7 y 9.8%, approx. equal for 1 and 2
18172030 multicenter, 11 Median age 33 y prevention
sites Results: 2 prevention: 44% Approp shocks: elevated EDP (HR:
Exclusion criteria: N/A Comps: total 30%, 5% early 1.3), nonsust VT (HR: 3.7)
Size: 121 Approp shocks: 30% Inappropriate shocks 5.8%/y
Annual rate approp: 1 7.7%, 2 9.8% (p=0.11) Comps 30%: 21% leads, 6%
generator
Zeppenfeld K et Study type: Inclusion criteria: 1 endpoint: Ablation of VT in congenital heart VT ablation of anatomic isthmus
al. Circ 2007 Single center repaired congenital disease successful: 91% without recurrence
(522) retrospective heart disease patients followup 30 mo during 30 mo followup
17967973 with sustained VT, Results: SR voltage map, identify scar: anatomic
Size: 11 undergoing voltage isthmus: between TV-RVOT, pulm annulus and RV
map, ablation free wall, pulm annulus and septal scar, septal scar
and TV
Exclusion criteria: N/A Ablation of isthmus (most common between TV
and anterior RVOT) abolished all 15 VT circuits.
van Zyl M et al. Study type: Inclusion criteria: 1 endpoint: outcome VT ablation in congenital VT ablation in ACDH: reentrant VT
HR 2016 (523) single center repaired congenital heart disease: SCD or appropriate ICD shock targets anatomic isthmus:
26961296 retrospective heart disease patients Mean followup 33 mo with confirmed block, no recurrent
with VT undergoing Results: Reentrant VT 67%, Focal 33% VT
Size: 21 ablation Isthmus dependent VT mechanism in 67%,
Mean age 45 y conduction block confirmed in 8
71% males

Exclusion criteria: N/A


Kapel GF et a. Study type: Inclusion criteria: TOF 1 endpoint: TOF VT ablation in LV outcomes TOF VT ablation in LV successful
CAE 2014 (524) Retrospective, 2 patients with VT in 4 patients: no recurrence during
25151630 centers ablation Results: Left sided mapping/ablation if right side 20 mos
RFA failed, part of circuit in LV Rt side failure: septal hypertrophy
Size: 28 Exclusion criteria: N/A 4/28 VT ablations used LV approach 2, pulmonary homograft 1, VSD
Target anatomic isthmus with transection patch 1

280

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Kapel GF, et al. Study type: 2 Inclusion criteria: 1 endpoint: Ablation of VT in CHD Predictors of lack of success:
Circ AE 2015 centers, repaired CHD pts followup 46 mo. 41% prior ICD No complete procedural success,
(525) retrospective undergoing ablation decreased LV function
25422392 Results: complete success 25/34 pts: 74%; 18/25 Transection of VT isthmus feasible
Size: 34 Mean age 48 y had preserved fxn in 74%
74% male Procedural failure: hypertrophy, pulm homograft,
TOF 82% prox to HBE, no critical reentry
TGA; VSD, AVSD, PS 79% discharged with ICD
Sustained VT 79% 15/18 complete success + preserved function d/c
on no AADno recurrences
Exclusion criteria: N/A 4 late deaths, 2 CHF, 2 CA
Kapel GF et al. Study type: Inclusion criteria: 1 endpoint: TOF VT isthmus identification TOF VT: slow conducting
EHJ 2017 (526) Single center repaired TOF patients anatomic isthmus is dominant
27233946 with VT Results: slow conducting anatomic isthmus substrate
Size: 74 induction/mapping identified by electroanatomical mapping: targeted
63% male for ablation
Mean age 40 y 28 patients with inducible VT. Ablation in 18 of
Exclusion criteria: N/A isthmus
Khairy P et al. Study type: Inclusion criteria: TGA 1 endpoint: TGA s/p atrial baffle ICD outcomes TGA s/p atrial baffle: ICD
CAE 2008 (527) Retrospective s/p atrial baffle with appropriate shocks mainly in
19808416 multicenter, 7 ICD Results: 2 prevention: 38% patients with 2 prevention, (HR:
sites Mean age 28 y, 89% Annual rates approp shocks: 18; p=0.034) and lack of BB, (HR:
male 1 0.5%, 2 6% 16.7; p=0.03)
Size: 37 Exclusion criteria: N/A Independent predictors: 2 prevention, lack of BB SVT preceded VT in 50% of
Approp shocks: None with inducible VT; approp shocks
37% of patients without inducible VT (p=0.043) Inducible VT did not predict
Comps 38%, 33% lead, 3% generator appropriate shock treatment in
TGA
Protective effect of BB
Tutarel O et al. Study type: Inclusion criteria: 1 endpoint: all-cause mortality ACHD 9-fold (864%) increase in ACHD
Eur H J 2014 retrospective ACHD patients 60 y at patients >60 y between 2000 and
(528) cohort, Royal entry, followed Results: 14.6% died (55/375) 2011
23882067 Brompton 1/2000-3/2012, mean Cardiac deaths: 40% CHF, CAD
age 65 y, median Independent predictors mortality: CAD (HR: 5.05);
Size: 375 followup 5.5 y CHF (HR: 2.36); NYHA class (HR: 1.96); mod-severe
systemic vent dysfunction (HR: 1.90)
Exclusion criteria: N/A
281

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Koyak Z et al. Study type: Inclusion criteria: 1 endpoint: SCD in ACHD Increased risk SCD: severe
Europace 2017 Multicenter ACHD; age matched ventricular dysfunction, increase
(529) case-control: controls; mean Results: 131 SCD, mean age 3614 y QRS duration 5 ms/y
27247006 CONCOR, followup 7 y Increased risk: increase in QRS duration 5 ms/y
Toronto, Leuven (OR: 1.9), change in systemic vent fxn to severe
Size: 25,000 Exclusion criteria: N/A (OR: 16.9; 95% CI: 1.8120.1, p=0.008)
Engelfriet P et Study type: Inclusion criteria: 1 endpoint: ACHD morbidity VEA highest in TOF 14%;
al. EHJ 2005 multicenter ACHD patients in Median followup 5 y Cyanotic 6%, VSD 3%,
(530) retrospective Europe: ASD, VSD, Results: Ventricular arrhythmias:
15996978 TOF, coA, TGA, TOF 14%, cyanotic 6%, VSD 3%, others 2% except
Size: 4110 Marfan, Fontan, Fontan: 0
cyanotic SVT: Fontan 45%, ASD 28%, TGA 26%, TOF 20%,
cyanotic 16%
Exclusion criteria: 8 Endocarditis: VSD 7%, cyanotic 6%, TOF 4%, others
lesions included 0-2%
Gallego P et al. Study type: Inclusion criteria: 936 1 endpoint: Causes SC arrest in ACHD Highest SCA:
AJC 2012 (531) single center ACHD patients TGA 10/1000
22464215 retrospective followed single center Results: SCA 2.6/1000 pt y UVH, coarctation, TOF
8387 patient-y of SCA occurred in 23% of severe subaortic Severe subaortic ventricular
Size: 22 followup ventricular dysfunction, vs 0.7% with nonsevere dysfunction (HR: 29)
dysfunction, p<0.001
Exclusion criteria: N/A 80% of SCA occurred in TGA, UVH, coarctation,
TOF
Engelings CC et Study type: Inclusion criteria: 1 endpoint: Identify cause of death in ACHD Leading causes of cardiac death:
al. Int J Cardiol National cohort ACHD patients >18 y, CHF 28%, Sudden 23%
2016 (532) mean followup 3.7 y; Results: 239 deaths, 9.2%, mean age 39.817.8 y Sudden death highest: Marfans,
26970963 Size: 2596 between 1/01-1/15 Related to Cong HD: 72%: CHF 28%, SCD 23% AS, Eisenmenger syndrome, cc TGA,
Leading causes: CHF-UVH, TGA TGA, TOF, VSD, UVH
Exclusion criteria: N/A SCD: Eisenmenger, TOF, Marfan, AS AICD under-utilized
Comparing 2001-2008 with 2009-2015:
CHF increased from 23-30%, SCD decreased from
29-20%

282

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Fish FA (533) Study type: Inclusion criteria: Use 1 endpoint: Adverse events during treatment Deaths 5.6%, CA 4.8%, pro-
JACC 1992 Retrospective of class Ic AA meds in with flecainide or encainide for VA: Pro- arrhythmia 6.4% for patients
1906902 multi-center 124/579 young arrhythmia, CA/SD treatment for VA with either
patients with VA flecainide or encainide
Size: 124 Flecainide 103, Results: Flecainide: Pro-arrhythmia: 5.8%, CA
(entire study, encainide 21 3.9%, sudden death4.9% for SVT patients, risk higher if
579) Encainide: pro-arrhythmia 9.5%, CA 9.5%, sudden structural HD, not for VT
Exclusion criteria: N/A death9.5%
Efficacy 71-76%
10 patients CA/Death: most on flecainide
Stan MN et al., Retrospective ACHD patients 1 endpoint: Identify incidence and risk factors Highest Risk: low BMI <21,
2014 (534) single center developing amio- amio cyanotic HD
22518347 induced thyrotoxicosis
23 after 3 mos amio, Results: Thyrotoxicosis13.6% (23/169) ACHD
Mayo Clinic 1987- patients developed amio thryrotoxicosis.
2009; median
followup3.1 yrs.

Silka MJ et al. Study type: Inclusion criteria: 1 endpoint: Population based risk of SCD in Late SCD: 4 lesions: 1/454
JACC 1998 (535) Retrospective congenital heart congenital heart disease patient-y
9669277 statewide disease surgery in Aortic stenosis
registry Oregon 1958-1996 Results: SCD 1/1118 patient-y Coarctation
3589 patients 37/41 late sudden deathoccurred in 4 lesions TGA
Size: 41 Causes SCD: arrhythmia 75%, CHF 10%, other TOF
Exclusion criteria: cardiac 17% (embolic, aneurysm rupture) Cause SCD: arrhythmia 75%, CHF
single ventricle not 10%
included
Oechslin EN et Study type: Inclusion criteria: 1 endpoint: Mortality causes in ACHD Highest mortality lesions
al. AJC 2000 single center ACHD patients Results: Mean age death 37 y congenital heart disease:
(536) retrospective followed Toronto, Causes: sudden 26%, CHF 21%, periop 18% univentricular 41%;
11074209 2609 adults Youngest age at death: TGA, tricuspid atresia, PA, ccTGA 26%,
Size: 197 aortic coarc <30 y TOF or PA 16%,
Exclusion criteria: N/A >50 y; ASD, PDA Ebstein 9%
AVSD 7%,
Nieminen HP et Study type: Inclusion criteria: 1 endpoint: Causes of death in ACHD during Causes of late death in congenital
al. JACC 2007 National Finland national 45 y followup heart disease: cardiac 67%: CHF
(537) registry of congenital
283

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
17888844 registry, heart disease, 6024 Results: 45 y survival 89%, lower than gen 40%, periop 26%, SCD 22% other
retrospective patients surviving first population CV 12%
operation Highest risk CD: TGA, UVH, TOF, VSD Highest risk of SCD: coA 42%, TOF
Size: 592 Other CVD: stroke, arrhythmia, pulm emboli, and TGA: 30%
Exclusion criteria: N/A endocarditis, aortic rupture Increased non-cardiac death 2
Increased non-cardiac mortality fold: neurologic, respiratory
Verheugt C et Study type: Inclusion criteria: 1 endpoint: Complications in ACHD Ventricular arrhythmias overall
al. IJC 2008 (538) Meta-analysis ASD, VSD, PS, TOF, 7%, highest TOF 14%
18687485 MEDLINE 1980- coarctation, TGA Results: Vent arrhythmias: TOF 14%, VSD 2.9%, MI highest coarctation 5%
2007 Exclusion criteria: TGA 1.9% SVT: all lesions: 18%
univentricular heart SVT: TGA 26%, ASD 28%TOF 20%
Size: 7894 Summarizes endocarditis, CHF, CVA, MI, SVT by
lesion
Pillutla P et al. Study type: Inclusion criteria: CDC 1 endpoint: ACHD death trends Decline in mortality among TGA,
AHJ 2009 (539) CDC registry registry 1979-2005, TOF
19853711 causes of death congenital heart Results: Cyanotic lesions: arrhythmia, then HF
disease in USA Non-cyanotic lesions, MI after 1990, arrhythmia MI leading cause of death in
Size: prior to 1990 patients with non=cyanotic lesions
Exclusion criteria: N/A
Verheugt CL et Study type: Inclusion criteria: 1 endpoint: ACHD causes of death Lesions with highest mortality:
al. EHJ 2010 Dutch CONCOR 6933 ACHD patients: Univentricular heart 25%,
(540) national registry, 197 deaths: 2.8% Results: Median age death 49 yrs DORV + TOF 13%
20207625 retrospective 77% CV cause: CHF 26% age 51 yrs, sudden ccTGA 6%
Exclusion criteria: N/A death19% age 38 yrs Ebstein 5%
Size: 197 Ventricular arrhythmias predicted SCD, HR 1.5 AVSD 5%
SVT and VT predicted CHF, HR 5.1 and 4.5 TGA 3%
See complications by lesion analysis!
Zomer AC et al. Study type: Inclusion criteria: 1 endpoint: ACHD causes of death SCD: 10% with exertion
IJC 2012 (541) Retrospective causes of death in Total followup 26,500 pt y Highest mortality: univentricular
20934226 national registry ACHD patients hearts 26%, TOF/DORV/PA 20%,
Results: Median age at death 48 y TGA and cc TGA 10%, AVSD 6%,
Size: 231 Exclusion criteria: N/A Causes of death: CHF 26%, SCD 22%, malignancy Ebstein 6%,
9%, pneumonia 4%
SCD exercise 8%,
Lower risk-ASD 3%, VSD 1.3%, AS 1%
Youngest age: TGA 33 y, AVSD 37 y, ASD age 61 y

284

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Diller GP et al. Study type: Inclusion criteria: 1 endpoint: Cause of death ACHD compared with Highest mortality: Eisenmenger,
Circ 2015 (542) Single center ACHD patients general age/gender matched, calculate SMR complex congenital heart disease,
26369353 cohort followed 1991-2013, (standardized mortality ratio) UVH
median followup 9.1 Results: 7.7% died, 0.72%/pt y SMR, p<0.001: Fontan: 23.4,
Size: 6969 yrs Leading causes: CHF 42%, pneumonia 10%, SCD Complex congenital heart disease
7%, cancer 6%, hemorrhage 5% 14.1,
Exclusion criteria: N/A SCD highest: TGA arterial switch 33%, AVSD 14%, Eisenmenger 12.8,
Fontan and single RV 13% each, complex systemic RV 4.9,
congenital heart disease 11%, Eisenmenger 9%, Ebstein 3.3,
TOF 6% TGA arterial switch 2.6 (0.08),
TOF 2.3, Marfan 2.2, coarctation
1.7
Raissadati A et Study type: Inclusion criteria: 1 endpoint: ACHD Late mortality causes Late 40 yr survival: simple defects
al. JACC 2016 Nationwide Patients undergoing 87%, complex 65%
(543) cohort study, cardiac surgery <15 y Results: early mortality 5.6%; late 10.4% 40 y freedom sudden death: 99%
27470457 Finland old between 1953- congenital heart disease related deaths: 6.6%: simple, 91% severe, (HR: 9.9)
2009 causes-CHF 28%, reop 14%, SCD 13%, other CV 8% Highest CV mortality: UVH, TGA,
Size: 10,964 Sudden deaths: arrhythmia/unknown 78%, MI 7%, TOF, VSD, coarc
Exclusion criteria: N/A aortic dissection 5% Increased lung, neuro, infectious
diseases
Sudden death ages: ASD 40 y, TOF 30 y, coarc 29 y,
Cancer higher than general population, especially
females, (RR: 5.9)
Teuwen CP et Study type: Inclusion criteria: 1 endpoint: ACHD Non-sustained VT: risk for Sustained VT/VF developed rarely
al. IJC 2016 (544) retrospective ACHD patients with sustained VT/VF in patients with only non-sust VT
26805391 cohort VA: Mean age 4014 y Recurrent sust VT/VF frequent in
Nonsust VT 71% patients presenting with sust VT/VF
Size: 145 Sustained VT 17% Results: 5/103 nonsust VT patients developed recommend wait and see
VF 12% sustained VT/VF approach for nonsust VT;
Exclusion criteria: N/A aggressive treatment for sust VT/VF

285

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Wells R et al. Study type: Inclusion criteria: Review side effects of chronic oral amio Patients with congenital heart
2009 (545) Retrospective ACHD, mean age 34.9 disease at higher risk for amio
19691680 multicenter y, receiving 36% developed thyroid dysfunction: 19 hyper, 14 adverse effects, esp women,
amiodarone for 6 mo; hypothyroid. Sig risk factors: Female gender (OR: cyanosis, Fontan, or dose >200 mg
Size: 20 patients case-control group. 3.0) cyanotic HD (OR: 7.0); Fontan (OR: 4.0);
Mean duration 3 y, dosage >200 mg/d (OR: 4.0)
mean dose 191 mg

Exclusion criteria: N/A


Afilalo J et al. Study type: Inclusion criteria: 1 endpoint: all-cause mortality ACHD Current ACHd populations
JACC 2011 (546) Quebec ACHD patients 65 y surviving to age 65 y or greater, co-
21939837 database 1993- old at entry, followed Results: most common types congenital heart morbid diseases most powerful
2005 up to 15 y disease: shunt lesions 60%, valvar 37%, severe 3% predictors of mortality; increased
Arrhythmias present: AF 25%, Ventricular CAD 7% vs 5% age matched
Size: 3239 Exclusion criteria: N/A arrhythmias 34% Ventricular arrhythmias present
Mortality driven by co-morbidity: dementia (HR: in 34%
3.24), GI bleed (HR: 2.79), chronic kidney disease Prevalence ACHD in geriatrics: 3.7
(HR: 2.5); CHF (HR: 1.98), diabetes (HR: 1.76), /1000 (vs 4.2/1000 in non-geriatric)
COPD (HR: 1.67)
El Malti R et al. Study type: Inclusion criteria: 1 endpoint: Screening congenital heart disease Familial AV block/ASD correlated
EJ Human retrospective familial congenital for FATA4, NKX2.5, ZIC3 with NKX2.5
Genetics 2016 heart disease genetic Can be used to screen high risk
(547) Size: 154 screening Results: 10.4% identified with causal gene SCD families
26014430 NKX2.5 identified in ASD/VSD and conduction
Exclusion criteria: N/A disorders; 6/154, 3.9%
ZIC3 1.9%, GATA4, 0.7%

Abou Hassan Study type: Inclusion criteria: 1 endpoint: Screening NKX 2.5 gene defect in Familial septal defects and
OK et al. Sci Rep retrospective congenital heart congenital heart disease conduction disorders: high
2015 (548) disease in Lebanon: prevalence NKX2.5, SCD
25742962 Size: 188 high incidence of Results: Familial ASD: 60% with NKX 2.5
cosanguinity Diversity of phenotypes: congenital heart disease,
AV block, SCD, coronary sinus disease
Exclusion criteria: N/A
Ellesoe SG et al. Study type: Inclusion criteria: 1 endpoint: NKX 2.5 occurrence in familial Screen familial ASD patients for
CHD 2016 (549) Probands with familial congenital heart disease NKX 2.5, esp if conduction
26679770 Size: 39 disorders
286

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
congenital heart Results: NKX 2.5 found 2.5% of probands
disease

Exclusion criteria: N/A


Cuypers JA et Study type: Inclusion criteria: ASD 1 endpoint: ASD surgical repair long-term Surgical repair ASD: late SCD 1.5%
al. Heart 2013 Longitudinal surgical repair 1968- outcomes
(550) cohort 1990 Mean Followup 35 y
23886606
Size: 135 Exclusion criteria: N/A Results: SVT: 16%, late SCD 1.5%
Pacemaker 6%.
LVEF 58%, RVEF 51%. Low RVEF 31%, dilated RV
20%
Kuijpers JM et Study type: Inclusion criteria: ASD 1 endpoint: ASD secundum outcomes: gender ASD secundum outcomes: males
al. EHJ 2015 Dutch national secundum in Dutch differences higher risk conduction
(551) registry registry Cumulative followup 13584 pt-y disturbances, SVT, CVA, CHF;
25883174 Mean age 45 y decreased life expectancy c/w
Size: 2207 Males 33% Results: Median survival: men 79.7 y, women 85.6 general population
y.
Exclusion criteria: N/A Compared w age/sex matched gen pop, survival
for males lower; equal for females.
Khairy P et al. Study type: Inclusion criteria: TOF 1 endpoint: TOF arrhythmia outcomes & TOF Ventricular arrhythmias 15%,
Circ 2010 (552) Retrospective repair correlates increased with LV diastolic
20713900 multi-center Female 54% dysfunction
Mean age 37 y Results: Sustained arrhythmia: 43%. AF and Vent arrhythmias
Size: 556 Exclusion criteria: N/A Prevalence AT 20%: RAE, HTN, number of surgeries increased after age 45 y

ventricular 14.6%: number of surgeries, QRS


duration, LV diastolic dysfunction (OR: 3.3)
Valente AM et Study type: Inclusion criteria: TOF 1 endpoint: TOF risk factors death, VT TOF predictors SCD, VT:
al. Heart 2014 Prospective adults RVH, ventricular dysfunction (RV or
(553) multi-center Median age 24 y Results: 3.7% death/VT, median age 38 y LV), and AT
24179163 INDICATOR Cos regression outcomes predictors:
cohort Exclusion criteria: N/A RV mass/volume ratio 0.3, (HR: 5.04) Higher RV systolic pressure, HR
LVEF z score <2, (HR: 3.34) 1.39
Size: 873 AT, (HR: 3.65)

287

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Arya S et al. Study type: Inclusion criteria: TOF 1 endpoint: TOF outcomes: risk changing? TOF late SCD: 1.8%
CHD 2014 (554) Retrospective Late followup
24314315 single center Male 49% Results: Arrhythmias 54%: older postop interval,
Ages 17-58 y wide QRS mean 158 msec.
Size: 109 No correlation with surgical era, gender RV
Exclusion criteria: N/A pressure, RVOT gradient, RVEDV
Wu MH et al. Study type: Inclusion criteria: TOF 1 endpoint: TOF late arrhythmia outcomes TOF tachycardia in adults: 6.6%:
HR 2015 (555) National repair Taiwan; VT 18%, VF 3%,
25461497 database Taiwan database those born Results: Prevalence TOF in adults 0.06/1000 Median age VT/VF 2325 y
retrospective 2000-2010 reviewed Survival 10 y: 78% Interventions for tachycardia
(national health for late outcomes Arrhythmias 4.6%: 73% tachycardia 2.4% annually, adults
insurance! Easily 58% males Overall tachycardia: 3.3% (6.6% adults, 1.8% peds).
accessible care!) AF 29%. AVB 0.6%
Exclusion criteria: N/A SVT/AT/AFL/AF = 80%, VT 18%, VF 3%
Size: 4781 Mortality with VT: 24%, VF 60%.
Heng EL et al. Study type: Inclusion criteria: TOF 1 endpoint: TOF outcomes and biomarkers TOF: BNP level 15 pmol/L
Heart 2015 (298) Single center patients with Median followup 10 y associated with 5 fold increased
25351509 prospective age/gender matched Measured aldosterone, ANP, BNP, renin, risk death
controls. endothelin Incorporate BNP into risk
Size: 90 stratification
BNP 1pmol/L = 3.472 Results: Late deaths: 9%
pg/ml BNP 15 pmol/L: increased mortality (HR: 5.4),
sustained VT, (HR: 2.06)
Exclusion criteria: N/A
Drago F et al. Study type: Inclusion criteria: 1 endpoint: TOF voltage mapping of ventricular TOF scar extension correlates
IJC 2016 (556) Retrospective endocardium with risk factors for life-threatening
27505328 single center Exclusion criteria: arrhythmias
Results: 97% with scar in RVOT.
Size: 146 Total scar extension c/w: QRS 180 ms, LV and RV
dysfunction, PVC, prior shunt, re-intervention,
duration of post surgical followup

Kriebel T et al. Study type: Inclusion criteria: 1 endpoint: TOF patients undergoing ablation, TOF VT Ablation acute success
JACC 2007 (557) single center repaired TOF patients contact mapping, RF ablation 100% (8 patients)
18036455 retrospective with VT undergoing Recurrence 25% in 35 mo
ablation Results: 13 VT circuits, 2 focal
Size: 10 Males 75%; Age 52 y ICD pre in 2, recommended post in all

288

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: N/A
Witte KK et al. Study type: Inclusion criteria: 1 endpoint: TOF patients with ICD vs dilated CM TOF patients: higher risk inapprop
Europace 2008 single center TOF patients with ICD shocks 25% vs 4%,
(558) retrospective compared with dilated Results: TOF appropr shocks 25%; inapprop 20% Death rate for TOF 5%, < DCM,
18442962 CM 21%
Size: 20
Exclusion criteria:
Lange R et al. Study type: Inclusion criteria: TGA 1 endpoint: TGA atrial switch outcomes. TGA atrial baffle risk factors SCD:
Circ 2006 (559) Single center with atrial repair: Mean followup 19 y Prior VSD closure, Mustard repair
17060385 retrospective Senning 79%
Mustard 21% Results: 25 y survival: Mustard 76%, Senning 91%
Size: 417 (p=0.002)
Exclusion criteria: N/A Mustard: die more often of arrhythmia (p<0.001),
reop baffles (p<0.0001);
Independent risk SCD: VSD closure (HR: 2.3),
Mustard (HR: 2.0)
Schwerzmann Study type: Inclusion criteria: TGA 1 endpoint: TGA s/p Mustard outcomes TGA s/p Mustard: late SCD or
M et al. EHJ 2009 Single center s/p Mustard repair Mean followup 9 y sustained VT: 9%
(560) retrospective Mean age 28 y QRS duration 140 msec highest
19465439 Results: Sustained VT/SCD 9%: risk factors: risk sVT/SCD (HR: 13.6; 95% CI: 2.9
Size: 149 Associated anatomic lesion (HR: 4.9), NYHA III 63.4)
Exclusion criteria: N/A (HR: 9.8), impaired subaortic RVEF (HR: 2.2)

AT 44%, not predictor of VT/SCD (HR: 2.7; 95% CI:


0.613)
Wheeler M et Study type: Inclusion criteria: TGA 1 endpoint: TGA atrial switch late outcomes TGA s/p atrial switch: 1
al. CHD 2014 Single center patients, s/p atrial Results: SCD 5.6% prevention ICD-no appropriate rx
(561) retrospective switch, Mustard or ICD 5.6% 1 prevention: no appropriate therapy Higher risk: older age at surgery,
24151816 Senning Patients with SCD: all with AT vs 29% AT in presence of AT, earlier era of
Size: 89 survivors surgery
Exclusion criteria: N/A

289

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Bouzeman A et Study type: Inclusion criteria: 1 endpoint: TGA atrial switch and ICD outcomes TGA atrial switch and ICD:
al. IJC 2014 (562) Retrospective TGA s/p atrial switch Median followup 19 mo 9% appropriate therapy (1 pt, 1
25499397 multicenter, with ICD Results: 2 prevention 33%; prevention, successful ATP without
Median age 34 y Implant: one death during DFT (8%) shock)
Size: 12 All patients with severe vent dysfunction; 54% complications: 27%
Exclusion criteria: N/A worsening CHF, 5/11 (45%) transplanted. HF determines outcomes
50% sustained AT during followup
Buber J et al. Study type: Inclusion criteria: TGA 1 endpoint: TGA s/p atrial switch: ICD outcomes AT most common cause for ICD
Europace 2016 Retrospective s/p atrial switch with Median followup 4 y shocks in 1 prevention TGA s/p
(563) single center ICD implanted for 1 atrial switch
26705566 prevention Results: EPS performed 72%: sust VT 54%, AFL NOT predictive: VT inducibility,
Size: 18 Median age 26 y 31%. VT inducibility did not predict appropriate QRS duration, age
shock. 50% complications
Exclusion criteria: N/A One pt received shock for VT; 39% for SVT,
Inappropriate shocks: 61%, mainly SVT/AFL
Backhoff D et Study type: Inclusion criteria: TGA 1 endpoint: TGA s/p atrial switch: ICD rx TGA s/p atrial switch: low rate of
al. PCE 2016 Retrospective s/p atrial switch with Median followup 4.8 y appropriate ICD shocks 9%
(564) multicenter, 4 ICD. <<<inapprop shocks 24%
27503213 German centers Median age 27 y, 85% Results: 2 prev 12%. AT main cause of inappropriate
male. Shocks: Approp 9%, inapprop 24% shocks
Size: 33 Annual incidence approp rx: 1.9%/pt/yr. Vigorous treatment of AT, careful
Inducible VT/VF: no approp shock ICD programming (inactivation VT
Exclusion criteria: N/A 2 prev: no approp shock zone, program VF zone 220-230
No predictors of approp rx bpm)
Complications 21%

Pundi KN et al. Study type: Inclusion criteria: 1 endpoint: Fontan arrhythmia outcomes Fontan late outcomes:
CHD 2016 (565) Retrospective Fontan patients 5% VT, 5% late SCD
27545004 single center operated at Mayo Results: Freedom from arrhythmia requiring
1973-2012, with treatment: 10 y: 71%; 20 y: 42%; 30 y 24%. Risk factors: arrhythmias (65%),
Size: 996 questionnaire sent AFL /AT 48%, AF 19%, SVT AC /AVN 4%, AVV replacement, post bypass
VT 5%, SND 13%. Fontan pressure >20 mm Hg
Exclusion criteria: Predictors arrhythmia: AP Fontan, age at surgery
arrhythmia prior to >16 y, AT postoperatively. Preop sinus rhythm was protective
Fontan surgery

290

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Sakamoto T et Study type: Inclusion criteria: 1 endpoint: Late outcomes Fontan Late SCD in Fontan: 10% overall
al. Asian CVTS Retrospective Fontan patients 20/40 (50%) died Timely conversion of AP Fontan,
2016 (566) single center operated 1974-1986 Results: Causes of death in 20 patients: CHF 30%, medication to decrease ventricular
27563102 SCD 20%, arrhythmia 20%, other 30% volume and pressure load needed
Size: 40 Surgery: AP 70%, RA-
RV 25%
Exclusion criteria: N/A
Alexander ME Study type: Inclusion criteria: 1 endpoint: Sustained VT inducibility in Positive V stim correlated
et al. JCE 1999 single center congenital heart congenital heart disease decreased survival (HR: 6),
(567) disease patients arrhythmic events (HR: 3)
10466482 Size: 130 undergoing V-stim Results: Sust VT inducible 25% Patients with documented
TOF 33%, TGA 25%, Non-sust VT 12%, AFL or SVT: 32% clinical VT: 33% negative V stim
LVOT lesions 12% frequent false negative
Median age 18 y

Exclusion criteria: N/A


Silka MJ et al. Study type: Inclusion criteria: 177 1 endpoint: ICD outcomes in younger patients Early ICD study: 2 prevention
Circ 1993 (568) Multicenter patients age <20 y Mean followup 2.6 y 86%
8443901 retrospective undergoing ICD; Results: 2: ACA 76%, refractory VT 10%. 1: 5 y survival: 85%
125 with data Syncope with HD and inducible sustained VT: 10% SCD free survival 5 yrs: 90%
Size: 125 available. Shocks: appropriate 68% of patients, inapprop
Mean age 14.5 y 20%. 5 late SCD.
Cardiomyopathy 54%, Predictors late mortality: abnormal vent fxn
electrical 26%,
congenital heart
disease 18%

Exclusion criteria: N/A


Berul CI et al. Study type: Inclusion criteria: 1 endpoint: ICD comps & therapies young ICD in young patients: high
JACC 2008 (569) Multicenter Pediatric and Mean followup 7.5 y inappropriate shocks 28% in
18436121 retrospective congenital heart Results: 2 prev 48% congenital heart disease
disease patients Comps: early 14%, late 29%, electrical storm 5% Complications 43%
Size: 443 receiving ICD in 4 Appropriate shocks 26%, inapprop 21%--higher in
centers 1992-2004 electrical disease (31%) vs cardiomyopathy (13%),
Median age 16 y; 69% congenital heart disease (28%)
structural HD: SCD 1%
TOF 19%, HCM 14%

291

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Electrical 31%
Exclusion criteria: N/A
Khanna AD et Study type: Inclusion criteria: 1 endpoint: ACHD patients with ICD outcomes Appropriate ICD shock more likely
al. AJC 2011 Retrospective ACHD patients with Mean followup 2.2 y in patients with elevated
(570) single center, ICD subpulmonary pressure
21684513 Mayo TOF 44% Results: 1 prevention 64%
cc-TGA 17% Approp shock 19%, inapprop 15%
Size: 73 Exclusion criteria: N/A
Koyak Z et al. Study type: Inclusion criteria: 1 endpoint: ACHD ICD approp shock risk score. Appropriate shocks for ACHD:
CAE 2012 (571) Multicenter ACHD patients Median followup 4.6 y 2 prevention, (HR: 3.6)
22095638 retrospective 10 receiving ICD Results: 2 prevention 50% CAD, (HR: 2.7), and symptomatic
centers Mean age 41 y Shocks: approp 29%, inapprop 30%, (SVT 69%) nonsust VT (HR: 9.1)
Netherlands, TOF 51%, Septal defect Comps 29% High morbidity with ICD
Belgium 20%, ccTGA 13% 63% underwent PES: 73% inducible sust VT/pmVT, No assoc between ICD treatment
VF: no difference in appropriate shocks: 33% with and QRS duration
Size: 136 induc VT, 32% w/out Inducible sustained VT did not
Exclusion criteria: N/A In 1 prev patients, univariable risks symptomatic correlate with appropr shock
nonsust VT HR: 8; 95% CI: 2.327.1, p=0.001 and TGA patients: appropriate
subpulmonary ventricular dysfunction, HR: 3.0; therapy: 29% 2 prev, 4.3% 1
95% CI: 1.212.6, p=0.02 TOF patients: not at higher risk
approp rx
Khairy P et al. PACES/HRS Expert Consensus Statement 1 endpoint:
HR 2014 (572) on recognition and management of
24814377 arrhythmias in ACHD Results:

292

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 55. Nonrandomized Trials, Observational Studies, and/or Registries of S-ICD - (Section 11.1)
Study Acronym; Study Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Type/Design; (P values; OR or RR; Comment(s)
Year Published Study Size & 95% CI)
Bardy et al. Study type: Inclusion criteria: Meeting 1 endpoint: Successful immediate In small, nonrandomized studies, an
2010 (573) Prospective non- class I, IIa, IIb criteria for an ICD conversion of 2 consecutive episodes entirely S-ICD consistently detected
20463331 randomized clinical of induced VF each with a single 65-j and converted VF induced during EP
trials (covered 4 Exclusion criteria: GFR <30 shock. testing.
trials) ml/min, need for The device also successfully detected
antibradycardia pacing, Hx of Results: and treated all 12 episodes of
Size: N=78 in VT at rates <170 bpm and Mean age of the 78 patients was spontaneous, sustained VT
temporary S-ICD documented VT known to be 6111 y
implantation for reliably terminated with ATP All 6 patients underwent successful
testing 4 electrode implantation of the S-ICD, and in all the
configurations and patients, defibrillation with 65-J
DFT testing; N=49 submaximal shocks was successful
in a trial that during 2 consecutive episodes of
compared the best induced VF. Of 18 induced VF episodes,
of the tested S-ICD all were successfully detected by the
in the first trial device. After 488 d of FU, there were
with a transvenous no complications.
ICD system, In the 4th trial, 53 patients were
comparing DFTs; evaluated for sensing and defibrillation
N=6 followed by during implantation. Of 137 episodes
N=55 in trials that of induced VF, 100% were detected by
tested permanent the S-ICD. After 10 mo of FU, 53 of 55
S-ICD implantation. patients were alive. Pocket infection
developed in 2 patients. 12 episodes of
VT in 3 patients were successfully
treated during followup
Olde Nordkamp Study type: Inclusion criteria: Class I or IIa 1 endpoint: Effectiveness and safety The S-ICD is effective at terminating
et al. 2012 (574) Retrospective indication for a 1 or 2 of the S-ICD VA
23062537 study prevention ICD Rate of inappropriate shocks was
Results: Mean age=50 y. After 18 mo 13%
Size: N=118 Exclusion criteria: None of followup, 8 patients experienced 45 The rate of complications decreased
successful appropriate shocks (98% with improved technology and
first shock conversion efficacy). No implanters experience.

293

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
sudden deaths occurred. Fifteen
patients (13%) received inappropriate
shocks, mainly due to T-wave
oversensing, which was mostly solved
by a software upgrade and changing
the sensing vector of the S-ICD. Sixteen
patients (14%) experienced
complications. Adverse events were
more frequent in the first 15
implantations/center compared with
subsequent implantations.
Kobe et al. 2013 Study type: Inclusion criteria: Patients 1 endpoint: Short and long term Failure of conversion of induced VF
(575) Retrospective with a 1 or 2 prevention effectiveness and safety with the S-ICD set to standard polarity
23032867 case-control study indication for an ICD was 10.4%, and there were comparable
(matching was Results: Conversion rates of induced inappropriate shock rates during short-
done on the basis Exclusion criteria: None VF were 89.5% with a 65J shock, and term follow-up.
of sex and age) mentioned 95.5% including reversed shock
polarity in the study group.
Size: N=138 Termination of induced VF was
successful in 90.8% of the control
patients (p=0.815). Procedural
complications were similar between
the 2 groups. During a mean follow-up
of 217 d, 3 patients with S-ICD were
appropriately treated for VA. Three
inappropriate shokcks (5.2%) occurred
in 3 S-ICD patients due to T-wave
oversensing, whereas AF with rapid
conduction was the predominant
reason for inappropriate therapy in
conventional devices (p=0.745).
de Bie et al. Study type: Inclusion criteria: All patients 1 endpoint: Suitability for an S-ICD After 5 y of follow-up, approximately:
2013 (576) Retrospective who received a single- or dual defined as not reaching one of the i. 55% of the patients would have
23704324 study chamber ICD in the Leiden following endpoints during follow-up: been suitable for an S-ICD.
University Medical Center (1) an atrial and/or right ventricular ii. Significant predictors of
Size: N=1,345 between 2002 and 2011. pacing indication, (2) successful anti- unsuitability for an S-ICD were: 2
tachycardia pacing without a

294

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria: Patients subsequent shock or (3) an upgrade to prevention, severe HF and
with a pre-existent indication a CRT-defibrilator device. prolonged QRS duration.
for cardiac pacing were iii. No mention of patients with ESRD
excluded. Results: During a median follow-up of (mean GFR 85-89 ml/min)
3.4y, 463 patients (34%) reached an
endpoint. The cumulative incidence of
ICD recipients suitable for an initial S-
ICD implantation was 55.5% after 5 y.
Appropriate ATP and the necessity of
cardiac pacing resulted in the
unsuitability for an S-ICD in
approximately 94% of the cases,
whereas device upgrade was
responsible for the unsuitability in
approximately 6% of the cases.
Weiss R. et. al Study type: Inclusion criteria: Adult 1 endpoint: The 180 d S-ICD system This study supports the efficacy and
2013 (577) Prospective non- patients with a standard complication-free rate compared with safety of the S-ICD System for the
23979626 randomized indication for an ICD. a pre-specified performance goal of treatment of life-threatening VA.
multicenter trial 79%.
Exclusion criteria: Patients The 1 effectiveness end point was the
Size: N=321 (314 who required pacing or had induced VF conversion rate compared
were implanted documented pace terminable with a pre-specified performance goal
successfully) VT. of 88%, with success defined as 2
consecutive VF conversions of 4
attempts.

Results: Followup was for 11 mo.


Mean age was 52 y. The 180 d system
complication-free rate was 99%, and
sensitivity analysis of the acute VF
conversion rate was >90% in the entire
cohort. There were 38 discrete
spontaneous episodes of VT/VF
recorded in 21 patients (6.7%), all of
which successfully converted. Forty-
one patients (13.1%) received an
inappropriate shock.

295

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
There were no cases of lead failures,
endocarditis or bacteremia,
tamponade, cardiac perforation,
pneumothorax, hemothorax, or
subclavian vein occlusion associated
with the S-ICD System. There was no
electrode or pulse generator
movement in 99% of implanted
patients throughout the followup
period.
Olde Nordkamp Study type: Inclusion criteria: Patients 1 endpoint: To determine the In patients without an indication for
et al. 2014 (578) Prospective non- more than 18 y old with a prior prevalence of patients who are not bradycardia- or resynchronization
24320684 randomized study ICD implantation visiting the suitable for pacing, 7.3% were not suitable for S-
ICD outpatient clinic. a S-ICD according to the QRS-T ICD implantation according to the QRS-
Size: N=230 morphology screening-ECG; (2) to T morphology screening-ECG. This
Exclusion criteria: Patients identify clinical characteristics of these indicates that this prerequisite
who were pacemaker- patients; and (3) to analyze whether screening method is not limiting S-ICD
dependent or had an standard 12-lead ECG parameters can selection for most patients.
indication for pacing during be used to predict QRS-T morphology
implantation (i.e., ICD settings screening failure.
other than VVI 40 or Patients were defined suitable when at
DDI 40). Also patients with an least 1 sensing vector was considered
indication for appropriate in both supine and
resynchronization standing position.
pacing.
Results:
In total, 7.4% of patients, who were all
male, were considered not suitable for
a S-ICD according to the QRS-T
morphology screening-ECG.
Independent predictors for TMS failure
were HCM (HCM; OR: 12.6), a heavy
weight (OR: 1.5), a prolonged QRS
duration (OR: 1.5) and a R:T ratio <3 in
the lead with the largest T wave on a
standard 12-lead surface ECG (OR:
14.6).

296

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Randles et al. Study type: Inclusion criteria: ICD patients 1 endpoint: S-ICD eligibility that About 85.2% of patients with an
2014 (579) Prospective non- with no ventricular pacing. required 2 leads to satisfy the S-ICD indication for a 1 or 2 prevention ICD
24351884 randomized study screening template in both erect and have a surface ECG that is suitable for
Exclusion criteria: Patients supine positions. S-ICD implantation when assessed with
Size: N=196 with an S-ICD, patients with a an S-ICD screening template. A
paced QRS complex, and Results: Overall, 85.2% of patients prolonged QRS duration was the only
patients who were unable to (95% CI: 80.290.2%) fulfilled surface baseline characteristic independently
stand for the time required to ECG screening criteria. associated with ineligibility for S-ICD
record an erect ECG. The proportion of patients with 3, 2, 1, implantation.
and 0 qualifying leads were 37.2%
(95% CI: 30.444.0%), 48.0% (95% CI:
41.055.0%), 11.2% (95% CI: 6.8
15.6%), and 3.6% (95% CI: 1.06.2%).
The S-ICD screening template was
satisfied more often by Lead III (1
vector, 83.7%, 95% CI: 78.588.9%)
and Lead II (2 vector, 82.7%, 95% CI:
77.488.0%) compared
with Lead I (alternate vector, 52.6%,
95% CI: 45.659.6%).
EFFORTLESS S- Study type: Inclusion criteria: Patients 1 endpoint: Effectiveness and safety This study showed appropriate
ICD Registry Prospective and receiving a S-ICD of the S-ICD. system performance with clinical event
Lambiase et al. retrospective rates and inappropriate shock rates
2014 (580) observational Exclusion criteria: Specific Results: Complication-free rates were comparable with those reported for
24670710 study contraindications include class 97 and 94%, at 30 d and 360 d, transvenous ICDs.
I indications for permanent respectively. 317 spontaneous
Size: N=472 (241 pacing, pace-terminable VT, episodes were recorded in 85 patients
studied and previously implanted during the follow-up period. Of these
prospectively) functional unipolar pacing episodes, 169 (53%) received therapy,
system. 93 for VT/VF. One patient died of
recurrent VF and severe bradycardia.
First shock conversion efficacy was
88% with 100% overall successful
clinical conversion after a maximum of
five shocks. The 360d inappropriate
shock rate was 7% with the vast
majority occurring for oversensing

297

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
(62/73 episodes), primarily of cardiac
signals (94% of oversensed episodes).
Groh et al. 2014 Study type: Inclusion criteria: Patients 1 endpoint: Rate of passing screening More work is needed on sensing
(581) Prospective non- who had previously undergone test and predictors of failure. algorithms on S-ICDs to increase pt
24755323 randomized study implantation of a transvenous eligibility for this device.
ICD for 1 or 2 prevention and Results: 8% of patients failed the
Size: N=100 who were not receiving screening test.
bradycardia pacing and did not Patients with T-wave inversions in the
have an indication for pacing inferior leads had a 45% chance of
were identified. failing the screening.

Exclusion criteria: See above.


EFFORTLESS/ Study type: Inclusion criteria: Patients 1 endpoint: Safety and effectiveness S-ICD demonstrated high efficacy for
IDE Registry Prospective and indicated for an ICD. of the S-ICD VT/VF. Complications and
Burke et al. retrospective inappropriate shock rates were
2015 (582) Exclusion criteria: Patients Results: Followup was for 651 d. reduced consistently with strategic
25908064 Size: N=882 (568 with recurrent VT reliably Spontaneous VT/VF events (N= 111) programming and as operator
from EFFORTLESS terminated with ATP and were treated in 59 patients; 100 experience increased.
and 308 from the patients in need of pacing. (90.1%) events were terminated with 1
IDE trials) Patients with ESRD were shock, and 109 events (98.2%) were
excluded from the IDE trials. terminated within the 5 available
shocks.
The estimated 3 y inappropriate shock
rate was 13.1%. Estimated 3 y, all-
cause mortality was 4.7% (95% CI:
0.9%8.5%), with 26 deaths (2.9%).
Device-related complications occurred
in 11.1% of patients at 3 y. There were
no electrode failures, and no S-ICD
related endocarditis or bacteremia
occurred. Three devices
(0.3%) were replaced for right
ventricular pacing. Themo
complication rate decreased by
quartile of enrollment
(Q1: 8.9%; Q4: 5.5%), and there was a
trend toward a reduction in

298

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
inappropriate shocks (Q1: 6.9% Q4:
4.5%).

Data Supplement 56. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries for WCD (Section 11.2)
Study Acronym; Author; Aim of Study; Patient Endpoint Results Summary/Conclusions
Year Published Study Type; Population (Absolute Event Rates, Comment(s)
Study Size (N) P values; OR or RR; &
95% CI)
Chung MK. Cardiol Clin. Review article N/A N/A Description of WCD indications, efficacy and
2014. (583) Study size: N/A limitations.
24793801
Chung MK, et al. J Am Study type: Inclusion 1 endpoint: Asystole was an important cause of mortality in
Coll Cardiol. 2010. (584) observational, post- criteria: All Observational study of SCA events.
20620738 market registry and patients compliance and Compliance was satisfactory with 90% wear time in
Social Security Death implanted and effectiveness >50% of patients and low sudden death mortality
Index signed consent during usage.
Size: 3569 post-market 80 sustained VT/VF events occurred in 59 patients
(1.7%). First shock success was 76/76 (100%) for
Exclusion unconscious VT/VF and 79/80 (99%) for all VT/VF. 8
criteria: N/A patients died after successful conversion of
unconscious VT/VF (survival 89.5% of VT/VF
events). Asystole occurred in 23 (17 died), PEA in 2
and respiratory arrest in 1 (3 died), representing
24.5% of SCA. During WCD use, 3541/3569 patients
(99.2%) survived overall. Survival occurred in 72/80
(90%) VT/VF events.
Survival was comparable to that of implantable ICD
patients.
Klein HU et al. Pacing Clin Review article N/A N/A Description of WCD indications, efficacy and
Electrophysiol. 2010. (585) Study size: N/A limitations.
19889186

299

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Data Supplement 57. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries for Special Considerations for Catheter
Ablation (Section 12)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Relevant 2 Endpoint (if any);
Author; Study Size (P values; OR or RR; Study Limitations;
Year Published & 95% CI) Adverse Events
Blanck et al. 1993 Study type: Inclusion criteria: Results: BBRVT typically occurs in patients
(170) Single Center Review All patients at single 45 of 48 patients had SHD with SHD from a variety of causes in
8269297 center with BBRVT SHD was NICM in 16 patients with prolonged HV
Size: 48 patients diagnosed at EPS between patients, Ischemic CM in 23 conduction intervals.
1980-1992 patients, VHD in 2 patients BBRVT is associated with aborted
Exlcusion Criteria: SCD, Syncope, and Palpitations
7) Typical RBBB or Mean LVEF=23.2% BBRVT is most commonly
LBBB QRS associated with a LBBB QRS
morphology Clinical Presentation morphology, and less commonly
during VT Aborted SCD in 26% with RBBB or Interfascicular QRS
8) QRS preceded by Syncope in 51% morphologies
His and Sustained palpitations in Catheter ablation targeting the
appropriate BB 10% RBB or LBB is highly effective and
potential associated with a low risk of serious
9) Stable HV, RB-V, Mean HV interval in sinus complications.
or LB-V interval 80.4 msec
10) Induction
dependent on HV QRS morphology in VT
delay LBBB in 46 patients
11) Termination by RBBB in 5 patients
block in HPS Interfascicular reentry in 2
12) Noninducibility patients
after RBB ablation
Catheter Ablation
Performed in 28 patients
targeting the RBB in 26
patients and LBB in 2
patients
Successful ablation of VT in
100%
No Complications observed.

300

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Lopera et al. 2004 Study type: Inclusion criteria: Results: BBRVT occurs in patients with
(173) Single Center Review His Bundle, LBB, or RBB HPS VT induced in 20 of 234 both NICM and ICM, usually with
15028072 potential closely consecutive patients impaired LVEF.
Size: 20 patients associated with QRS with referred for VT ablation BBRVT is most commonly
any of associated with a LBBB QRS
the following: NICM: 9 of 81 patients morphology, and less commonly
4) H-H interval (11%) had HPS VT with RBBB or Interfascicular QRS
variation ICM: 11 of 153 patients morphologies
preceding similar (7.1%) had HPS VT Catheter ablation targeting the
V-V interval Mean LVEF 29+17% RBB or LBB is highly effective and
variation; 2 of 20 patients had normal associated with a low risk of serious
5) Anterograde LVEF complications if only one BB is
activation of the targeted and a higher risk of AV
bundle branches Clinical Presentation block if both BBs are targeted for
during ICD Shocks in 10 patients ablation.
tachycardia; or, Syncope in 3 patients
6) Abolition of VT by Other symptoms in 7
bundle branch patients
ablation.
Typical BBRVT in 16 of 20
Exclusion criteria: None patients
(all had LBBB QRS
morphology)
13 of 16 patients BBRVT
successfully ablated by RBB
ablation and 3 of 16 by LBB
ablation.
HV interval prolonged from
70+5.9 msec to 83+17 msec
after ablation.

Typical BBRVT and


Interfascicular VT in 2 of 20
patients. Ablation of both
the RBB and portion of LBB
eliminated VT in both

301

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
patients, complicated by AV
block in 1 pt.

Focal Mechanism from BBs


in 2 patients, one in RBB,
one in LBB. Ablation
eliminated focal VT in both
patients, complicated by AV
block in 1 pt.
Mehdirad et Study type: Inclusion criteria: Results: Catheter ablation of the RBB is
al.1995 (174) Single Center Review All patients undergoing RF HV interval 68+8 msec at effective for the treatment of
8771124 catheter ablation of the baseline BBRVT
Size: 16 patients RBB for BBRVT LVEF mean 31+15% BBRVT is associated with
prolonged HV conduction intervals.
RBBB developed in 15/16 The medium-term follow-up after
patients after RBB ablation catheter ablation of the RBB is
AV block occurred in 1 pt overall quite good.
After mean of 19+10 mo,
one patient died suddenly,
2 received cardiac
transplantation, and 1 died
of CHF.

HELP-VT Aim: Inclusion criteria: 1 endpoint: At 1y follow- Complications


Dinov 2014 (175) To determine the outcome of Patients with SHD referred up, VT free survival was Complications occurred in 11.1% of
24211823 VT catheter ablation in for catheter ablation of VT 57% for ischemic NICM and 11.1% of ischemic
patients with NICM to those with either NICM (N=63) cardiomyopathy and 40.5% cardiomyopathy patients, including
with ischemic cardiomyopathy or ischemic CM (N=164) for NICM patients (HR: 1.62; death in 4.8% of NICM and 3.7% of
Study type: Exclusion criteria: 95% CI: 1.122.34, p=0.01). ischemic cardiomyopathy
Prospective, non-randomized Failure of informed ischemic cardiomyopathy
Size: 227 patients consent required epicardial ablation
Intervention: in only 2 of 164 (1.2%)
Catheter ablation for whereas NICM required
patients with NICM

302

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Comparator: epicardial ablation in 30.8%
Catheter ablation in (p=0.0001).
patients with ischemic
cardiomyopathy

Euro-VT Study Aim Inclusion Criteria 1 Endpoint Complications


Tanner H 2010 To determine the safety and Drug and device Acute success with ablation Major complications occurred in
(176) efficacy of electroanatomic refractory, recurrent was achieved in 83% of 1.5% and minor complications in 5%
9656251 mapping and irrigated RF sustained VT after MI. mappable VTs and 40% of of patients, particularly groin
catheter ablation for VT after >4 episodes of sustained non-mappable VTs hematomas, with no procedural
MI VT in prior 6 mo. (p<0.0001). deaths.

Study Type: Exclusion Criteria During 12 mo follow-up, VT


Multicenter, non-randomized Age <18 y recurred in 49% of patients.
MI within 2 mo
Study Size LV Thrombus The mean number of
63 patients Unstable Angina therapies dropped from
Severe AS or MR 6070 prior to ablation to
Unwillingness to 1415 in the same period of
participate time (6 mo) after ablation
Intervention (p=0.02).
Electroanatomic mapping
and ablation with open-tip
irrigated catheter.
Post-approval Aim Inclusion Criteria 1 Endpoint Comments
Thermocool Trial To evaluate long-term safety Patient with coronary At 6 mo: 62% without VT Reduction in amiodarone usage and
Marchlinski F 2016 and effectiveness of RF disease, age 18 y and LV recurrence, proportion of hospitalization
(177) catheter ablation for VT in EF 10% with recurrent VT patients with ICD shock
26868693 patients with coronary disease (either 4 episode reduced from 81.2 (pre) to Improvement in QoL
documented by ICD, 2 26.8% and 50% reduction
Study Type: episode documented by in VT episodes in 63.8% of
Multicenter, non-randomized ECG in patients without patients.
ICD, incessant VT or
Study Size: 249 patients symptomatic VT despite Safety Endpoint
AAD treatment CV specific AE in 3.9% with
no stroke
Exclusion Criteria

303

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Mobile LV thrombus, MI
within 3 mo, idiopathic VT,
class IV HF, creatinine
2.5, recent cardiac
surgery, unstable angina,
severe AS or MR
Intervention
Electroanatomic mapping
and ablation with open-tip
irrigated catheter.

Data Supplement 58. Nonrandomized Trials, Observational Studies, and/or Registries Related to Post-Mortem Evaluation of SCD - (Section
13)
Study Acronym; Study 1 Endpoint and Results
Summary/Conclusion
Author; Type/Design; Patient Population (P values; OR or RR;
Comment(s)
Year Published Study Size & 95% CI)
de Noronha et Study type: Inclusion criteria: SCD 1 endpoint: Determine cause of SCD and The specialized cardiac
al. 2014 (586) consecutive cases referred by general compare initial diagnosis with that determined pathology exam appears to have
24148315 prospective pathologist to specialized at specialized center. value for determining specific
observational cardiac pathology center; Results: Data were skewed by age (median 32 causes of SCD in this population.
study SCD defined as witnessed y, range 1-98 y, 58% <35 y. Approximately 1/3 of Referring pathologists tended
SCA or unwitnessed SCD in the cases had a "cardiomyopathy", including to have a more difficult time
Size: 720 an individual alive and well idiopathic LVH (26%), HCM (20%) and ARVC identifying anatomically normal
up to 24 hs prior; non- (14%), and a category of obesity CM (14%) hearts, and over-diagnoses
cardiac causes ecluded at Coronary artery abnormalities accounted for cardiomyopathies.
initial autopsy 10%, with 79% of those being ASHD. In a The etiological data are not
comparison of diagnoses of 200 autopsies generalizable to the overall
Exclusion criteria: examined after referral, a disparity in final population because of skewing of
Non-sudden death; diagnosis was observed in 41% of the cases. A age at time of SCD for specialized
sudden-death in the misdiagnosis of cardiomyopathy was reported in cardiac evaluation.
context of worsening CHF; 37% referred cases, ultimately determined to
absence of age, sex, and have to be structurally normal.
circumstances of death

304

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Wu et al. 2016 Study type: Inclusion criteria: Deaths 1 endpoint: Causes of SCD, sub-grouped The proportion of SCDs that
(587) Retrospective that occur within 1h of the according to circumstances, sex and age groups were autopsy negative was
26844513 observational sudden loss of Results: strongly age-dependent, as was
cohort study of consciousness due to The peak incidence occurred between the ages the common autopsy-provable
anatomic and various CVD, or during of 31 and 60, with a 5-7-fold excess of causes.
histopathological sleep or unwitnessed, in males/females in that age range. Both The proportion of SCDs
findings in SCD which the affected persons incidence and male preponderance markedly attributed to dilated
victims between were considered healthy decreased in younger and older age groups. cardiomyopathy was surprisingly
1998 and 2013 24h before the event. Overall, 42% were due to CAD, 12% viral low, especially in the age group
myocarditis, and 5% cardiomyopathy, with 15% older than 35 y.
Size: 1656 SCD Exclusion criteria: Deaths being unexplained by autopsy. In age group
identified from a due to non-cardiac <35, CAD was 17% of cases, viral myocarditis
total of 3770 conditions, such as 27%, and unexplained 32%. At age >55, CAD
sudden deaths injuries, poisonings, accounted for 86%, viral <2%, and unexplained
(43.9%) from all epilepsy, acute pulmonary <1%.
causes during embolisms, and allergies.
the study period
Vassalini et al. Study type: Inclusion criteria: SCD in 1 endpoint: Clinical and postmortem findings Although this is a small study,
2016 (588) Retrospective subjects aged 1-40 y. of patients who died suddenly without a Hx of the exclusion of a prior Hx of
25575272 cohort autopsy prior heart disease. heart disease restricts this study
study Exclusion criteria: Prior Results: Coronary artery abnormalities in to SCD that occurred as a first
Hx of heart disease; 18.5% (including one with an anomalous cardiac event.
Size: 54 sudden infant death coronary artery origin); ARVD/C in 11.1%; LVH in One important finding is the
syndromes (under 1 y of 5 cases (9.2%), 3 of whom had myocyte association of SCD with the only
age), extracardiac causes disarray; VHD in 7.4%; myocarditis in 7.4%; abnormalities at postmortem
at autopsy; drug or alcohol pathological changes in the specialized found in the specialized
abuse found at conducting system in 22.2%, in the absence of conducting system in 22.2%
postmortem toxicology. any other anatomic or histopathological A second is the autopsy being
findings; in 12 cases (22.2%), autopsy was completely negative in another
completely negative in 22.2%. No postmortem 22.2%. No postmortem genetics
genetics done in this group were done in this subgroup
Tester et al. Study type: Inclusion criteria: 1 endpoint: Identification of SUD-associated Molecular autopsy provides a
2012 (589) Prospective Autopsy-negative SUDs variants in KCNQ1, KCNH2, SCN5A, KCNE1, reasonable yield of putative SUD-
22677073 cohort study referred for molecular KCNE2, or RYR2. associated variants, recognizing
autopsy. Candidate genes that the candidate genes were
Size: 173 restricted to KCNQ1, Results: Pathogenic mutations were identified restricted to the common LQTS-
KCNH2, SCN5A, KCNE1, in 45 autopsy-negative SUD cases (26.0%). LQT
305

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
KCNE2, and RYR2. SUD- variants more likely to be associated with SUD associated genes and the most
associated variants had to during sleep; CPVT (RyR2) more like associated common CPVT-associated gene.
be nonsynonymous, with SUD during exercise. Family Hx of SCD It is likely that broader panels,
involve a highly conserved positive among relatives of 11 of 45 variant- including other genetic disorders,
residue, and absent from positive probands. including structural disorders that
reference normal may not be identified on routine
populations autopsy, would increase this
yield.
Exclusion criteria: A prior
documented Hx of a
channelopathy in either
probands or family
members (Exception:
History of long QT on an
ECG mentioned in
autopsy)
Tang et al. Study type: Inclusion criteria: N/A 1 endpoint: N/A Comprehensive review on
2014 (590) Review article on postmortem molecular studies of
24157219 molecular Exclusion criteria: N/A Results: N/A SUD and autopsy-defined
diagnostic structural genetic disorders
protocol for SCD
Size: N/A
Papadakis et al. Study type: Inclusion criteria: Family 1 endpoint: Identification of genetic variants Victims of SCD with structural
2013 (591) Retrospective members of SCD probands associated with inherited arrhythmia syndrome findings of uncertain significance
23671135 cohort study, who died suddenly and in >1 relative(s) of probands who had structural are as likely to have genetic
with prospective had been apparently findings of uncertain significance (such as variants associated with inherited
cardiogenetic healthy, death from ventricular hypertrophy, myocardial fibrosis, arrhythmia syndromes as are
evaluation of natural causes, last seen and minor CAD). Comparison group was the those with normal autopsies.
family members. alive and well within 12 h, cohort of 163 families in whom the findings Findings call for caution in
with autopsy findings were consistent with SUD based on normal interpreting uncertain structural
Size: 340 showing structural autopsy. findings, with particular regard to
families abnormalities of uncertain Results: implications for family members
causal effect (e.g., 51% of the study group had genetic variants of probands.
ventricular hypertrophy, associated with SADS; for the comparison
myocardial fibrosis, or group, consistent with SADS, the proportion
minor CAD (N=41). with positive genetic findings was 47%.

306

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Exclusion criteria:
Incomplete postmortem
report, presence of an
extracardiac cause of
death, or positive
toxicology screen.
Harmon et al. Study type: Inclusion criteria: 36 of 45 1 endpoint: Autopsy-defined cause of SCD The adjudicated diagnosis
2014 (592) Cohort study athlete SCDs with agreed with the official pathology
24585715 from NCAA sufficient autopsy Results: report in only 59% of cases.
registry of information Autopsy-negative SUD in 11 (31%); coronary Autopsy-negative SUD was
athletes who artery abnormalities in 5 (14%), dilated CM in 3 common (31%)
died suddenly (8%), myocarditis in 3 (8%), aortic dissection in 3
Exclusion criteria: N/A (8%), and idiopathic LVH (possible HCM) in 3
Size: (8%). There was 1 case each (3%) of HCM, ARVC,
45 LQTS, commotio cordis, commotio cordis, and
Kawasaki disease. There was 1 case of death in a
sickle cell positive athlete who also had LVH.
There was 1 case of death in a sickle cell positive
athlete who also had LVH.
Bagnall et al. Study type: Inclusion criteria: SUD in 1 endpoint: Comparison of the yield of whole Study suggests the WES
2014 (593) Retrospective the 140 y age group, exome sequencingto common candidate gene increases the yield of molecular
24440382 analysis of de- classified as SUD based sequencing for identifying a potentially relevant autopsy in SUD by as much as 3-
identified cases upon sudden unexpected variant associated with autopsy-negative SUDs fold, compared to common
of autopsy- death with a negative in a population age 140 y. candidate genes for LQTS and
negative SUDs autopsy. Results: Based upon likely variants identified by CPVT.
WES, the yield increased from approximately Nonetheless, the majority of
Size: Exclusion criteria: 10% of cases to as much as 30%. molecular autopsies still fail to
28 Previous Hx of systemic identify a highly-likely or known
disease or alternative disease-causing mutation.
cause of death identified
after a complete autopsy,
including histopathologic
and toxicologic analysis
Anderson et al. Study type: Inclusion criteria: Stored 1 endpoint: Putative variants identified by There appears to be added
2016 (449) Whole exome DNA from SUD victims WES, excluding the previously studied common valve to WES, compared to a
27114410 sequencing of with previous negative candidate genes. limited candidate gene approach
stored DNA from molecular autopsies

307

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
referred cases of (21/32, 66%) using a Results: WES increased the yield compared to for molecular autopsies following
SUDY with common candidate gene the candidate genes, to 44% from 34%. SUD.
negative protocol (KCNQ1, KCNH2, Whether a broader candidate
autopsies SCN5A, RYR2) gene panel might achieve the
Size: same yield requires further study.
32 Exclusion criteria: The data suggest that the yield
Previous identification of a from WES is greater for the age
putativelt significant group 1-10 y, compared to 11-19
variant in KCNQ1, KCNH2, y, but this is not conclusive based
SCN5A, or RYR2 (11/32, upon the small numbers.
34%)
Bagnall et al. Study type: Inclusion criteria: 292 1 endpoint: Identification of relevant genetic 40% of SCDs in children,
2016 (594) Prospective, subjects with clinical and variants among subjects without autopsy or adolescents and young adults are
27332903 population- autopsy confirmed causes clinical identification of cause of SCD. classified as unidentified causes
based, clinical, of SCD (60%), and 198 based on autopsy and clinical
toxicological, (40%) subjects without Results: Among the total cohort, 292 subjects information.
autopsy, and identified cause based on had clinical and/or autopsy identified causes of In the age group 3035 y, a
genetic study of clinical or autopsy SCD (60%). The most common identified causes greater proportion of causes are
sudden cardiac information, among whom were CAD (24%) and inherited identified, and CAD is the
death among 113 underwent genetic cardiomyopathies (16%), while unexplained SCD dominant cause.
children and testing. accounted for 40% overall (N=198). Based on a partial sample of
young adults, cases with unidentified causes
age 135 y. Exclusion criteria: De- Among the 113 of 198 unexplained cases that that underwent post-mortem
identified cases; DNA had post-mortem genetic testing, 31 (27%) were genetic testing, an estimated 27%
Size: unavailable identified as having a clinically genetic variant. of such cases yielded evidence of
490 a clinically relevant genetic
variant.

Data Supplement 59. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries of Terminal Care - (Section 14)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Hill et al. 2015(595) Study type: Systematic Inclusion criteria: 1 endpoint: N/A concept Three broad themes
25239128 narrative review of Empirical studies mapping was performed for (1) Diverse preferences regarding
published in English discussion and deactivation.

308

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
published studies language between 2008 emergent themes from the (2) Ethical and legal considerations were
(2008 2014) and 2014, primarily set of studies predominant in Canadian and American
related to adults (above literature. Advance directives were
Aim: to evaluate the 18y) with an implanted Results: See conclusions uncommon in Europe.
evidence on patients' ICD and primarily related (3) Living in the now was evident among
perception of to the deactivation of patients.
implantable ICDs at end of life
cardioverter
defibrillator
deactivation at end of
life.

Size: N=18 studies

Lewis et al. 2014 (37) Study type: Inclusion criteria: 1 endpoint: N/A A significant degree of
24668214 Integrative review original quantitative and integrative review misunderstanding and inaccurate recall
qualitative research of information regarding ICD function at
Aim: To explore articles that directly Results: See conclusions. all decision
patients decision- studied the patient In terms of deactivation decisions, the
making experiences response regarding ICD majority of patients were not aware of
regarding ICDs from decision-making. this option.
the decision to 18 y of age orolder,
implant to the
consideration of Exclusion criteria articles
deactivation at end of that did not incorporate
life. the patients perspective,
if they
Size: N=25 studies solely focused on living
with or adjusting to the
ICD.
Kramer et al. 2016 Study type: Inclusion Criteria: 1 endpoint: Descriptive Half of patients over age 65 y dont
(596) Retrospective cohort Patients >65 y who had survive 5 y.
27016104 study (NCDR linked to ICDs inserted between Results: 1/3 of the decedents utilize hospice
Medicare) January 1, 2006 through 5 y after device implantation, services.
March 31, 2010 50.9% of patients were either
Aim: to describe the deceased or in hospice.
incidence and features Exclusion criteria:

309

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
of hospice use in a Not fee-for-service Among decedents, 36.8%
large, nationally Medicare patients. received hospice services.
representative sample Patients enrolled in Factors most strongly
of older patients hospice before device associated with shorter time
following ICD placement. to hospice enrollment were:
implantation, and to older age HR: 1.77;
identify factors class IV HF HR: 1.79;
associated with EF <20% HR: 1.57
hospice enrollment in Greater regional hospice use
this cohort.

Size: N=194,969

Buchhalter et al. 2014 Study type: Inclusion criteria: 1 endpoint: Descriptive Patients have deactivation decisions
(597) retrospective chart Patients with ICD referred very close to delay (median 2 d)
24276835 review Mayo clinic to the cardiac service for Results: Over half the time, this decision falls to
deactivation. 150 patients who had their a surrogate.
Aim: To describe ICD deactivated. Devices were not mentioned in
features and Exclusion criteria N/A Median of 2 d between advance directives.
outcomes of patients deactivation and death.
who underwent ICD Advance directives were
deactivation. present for 85 (57%) of these
patients, but only 1 of these
Size: N=150 made any mention of the ICD.
6 of the ICD deactivations
were for pacemaker-
dependent patients,
Surprisingly, surrogates were
responsible for over half
(51%) of the deactivation
decisions.
Palliative care consultation
was obtained in 43% of
patients.

310

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Goldstein et al. 2004 Study type: Telephone Inclusion criteria: 1 endpoint: Descriptive Deactivation discussions were not
(598) survey with next-of- Deceased patients: common and occurred late in the illness
15583224 kin of deceased median age 76 y at death; Results:
patients 27% women; 27% of next of kin recalled a Limitations
median implant time 27 discussion regarding 12 y old
Aim: To describe the mo. deactivation of the ICD with Relied on reports from the next-of-kin
frequency, timing, and their clinician. Recall bias (interviews occurred a
correlates of ICD Interviewed next-of-kin: 21% chose to deactivate. median of 2.3 y after patient death)
deactivation median age 67; These discussions all took
discussions majority were spouses. place in the last few d or h of
the patient's life.
Size: 100 27 patients received shocks in
the last mo of life,
8 patients received a shock
from their ICD in the min
before death.
Goldstein et al. 2010 Study type: Inclusion criteria: 1 endpoint: Descriptive Over half of hospices had had a patient
(599) Nationwide survey of Hospice directors get shocked by their ICD in the year prior
20194235 hospice providers (nursing, clinician, or Results: to their death.
administrative) 97% of hospices admitted
Aim: To determine patients with ICDs Older survey: more hospices have a
whether hospices are 58% reported that in the past policy now.
admitting patients year, a patient had been
with ICDs, whether shocked.
such patients are Only 10% of hospices had a
receiving shocks, and policy that addressed
how hospices manage deactivation.
ICDs. On average, 42% (95% CI, 37%
to 48%) of patients with ICDs
Size: 414 had the shocking function
deactivated.

311

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Berger et al. 2006 Study type: Inclusion criteria: 36/57 did not have Patients infrequently consider
(600) self-administered Patients with ICDs preferences for disabling. deactivation and rarely consider them in
16689116 survey 21/57 described situations in advance directives
Exclusion criteria: N/A which they would want
Aim: To assess deactivation. Limitations:
whether ICD recipients Advanced directives were Retrospective
have considered prepared by 35/57 subjects, Selection bias
preferences for none addressed the ICD.
disabling the ICD.

Size: N=57

Dodson et al. 2013 Study type: telephone Inclusion criteria: Following an informational Patients endorse preferences for ICD
(601) survey. Patients with ICDs, >50 y, script regarding the benefits deactivation in hypothetical scenarios
23358714 English speaking and harms of ICD therapy,
Aim: To examine 67/95 (71%) subjects wanted Limitations:
preferences for ICD Exclusion criteria: N/A ICD deactivation in 1 or more Single center
deactivation in scenarios.
hypothetical scenarios

Size:
N=95.
Goldstein et al. 2008 Study type: Inclusion criteria: No participant had ever Patients did not consider and had some
(602) Qualitative focus Patients with ICDs discussed deactivation with confusion about ICD deactivation
18095037 groups. their physician, nor knew that
deactivation was an option. Limitations:
Aim: To identify Some subjects expressed that Single center
barriers to ICD the physician should make the Small sample size
deactivation decision.
discussions in patients
with advanced illness.

Size: N=15
Habal et al. 2011 (603) Study type: semi- Inclusion criteria: Focused on subset of patients Patients expressed varied impressions
21514785 structured survey N=41 total patients with ICDs about deactivation
study N=19 with ICD 2/19 (11%) reported
discussing the possibility of Limitations:

312

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Aim: To determine HF ICD deactivation with their Convenience sampling
patients' awareness, physician. Single center
comprehension and Following clarification, 9/19 Small sample size
utilization of advanced (47%) stated they would want
care directives their ICD turned off should
their condition deteriorate.
Size: 41 (19 with ICDs) 5/19 (26%) would not want it
deactivated.
Kirkpatrick et al. 2012 Study type: Non- 1 endpoint: Descriptive Majority of patients are not addressing
(604) experimental, 30% women; their ICD in advance directives.
21943937 descriptive, telephone 85% Caucasian; Results: Patients want their doctors to have the
survey. median age 61 y; 140 subjects either had a conversation about deactivation.
mean implant time 61 living will or a power of
Aim: To explore mo; attorney. Limitations:
patients' preferences 100% 2 education and Only 3 (2%) of these subjects Study objectives not explicitly stated
for ICD deactivation in higher; included a plan for their ICD. Single center
the setting of a do not 38% with prior shock(s); 96% had never discussed what
resuscitate order mean number of shocks to do with their ICD at end-of-
and/or admission to 4.69. life with a medical
hospice. professional.
Nearly all wanted their
Size: N=278 physician to bring up the topic
of deactivation.
Kramer et al. 2011 Study type: Inclusion criteria: 1 endpoint: Descriptive Legality of ICD deactivation is not well-
(605) Non-experimental, Members of Hypertrophic known among patients
21296323 descriptive, online Cardiomyopathy Results:
survey. Association Widespread uncertainty and
confusion regarding the legal
Aim: To identify the status on implantable cardiac
ethical beliefs and device deactivation was
legal knowledge of found.
patients with HCM 57% were unsure if ICD
relating to end-of-life deactivation was legal.
care and the 198 patients with an ICD had
withdrawal of advanced directives, and only
implantable cardiac 15 (8%) specifically addressed
device therapy. their ICD.

313

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: N=546

Data Supplement 60. Nonrandomized Trials, Observational Studies, Guidelines, and/or Registries for Shared Decision Making (Section 15)
Study Acronym; Study Type/Design; Patient Population 1 Endpoint and Results Summary/Conclusion
Author; Study Size (P values; OR or RR; Comment(s)
Year Published & 95% CI)
Lewis et al. 2014 (606) Study type: Inclusion criteria: 1 endpoint: N/A integrative review A significant degree of
24668214 Integrative review Original quantitative and misunderstanding and inaccurate
qualitative research Results: See conclusions recall of information regarding
Aim: To explore articles that directly ICD function at all decision
patients decision- studied the patient points.
making experiences response regarding ICD The majority of patients were
regarding ICDs from decision-making. not aware of deactivation.
the decision to age 18y The desire to live trumped
implant to the inconveniences for most patients
consideration of Exclusion criteria articles but this appeared to be a
deactivation at end of that did not incorporate function of health state.
life. the patients perspective,
if they
Size: 25 studies solely focused on living
with or adjusting to the
ICD.
Dodson et al. 2013 Study type: telephone Inclusion criteria: Following an informational script Patients endorse preferences
(601) survey. Patients with ICDs, age regarding the benefits and harms of for ICD deactivation in
23358714 >50 y, English speaking ICD therapy, 67/95 (71%) subjects hypothetical scenarios
Aim: To examine wanted ICD deactivation in 1 or more Limitations: Single center
preferences for ICD Exclusion criteria: N/A scenarios.
deactivation in
hypothetical scenarios

Size: N=95.
Lewis et al. 2014 (607) Study type: mailed Inclusion criteria: 1 endpoint: 55 of 106 patients Over half of patients were
25070249 survey Adult patients with ICDs (51.9%) were unaware that ICD unaware that there was an

314

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
generator replacement was not option to not replace the ICD and
Aim: To assess patient Exclusion criteria: compulsory. a portion of them would have
awareness that ICD CRT considered it.
generator Results: Limitations: Single center and
replacement is If given the option, 15 of 55 (27.2%) Recall bias
optional, to gauge stated that they would have
their understanding of considered nonreplacement.
the risks and benefits For 88 of 106 patients (83.0%), it was
of ICD replacement, important or very important to
and to gain insight into discuss risks and benefits of continued
their decision-making therapy before deciding.
process.

Size: N=106 (response


rate 72%).
Hauptman et al. 2013 Study type: Focus Inclusion criteria: 1 endpoint: Patient focus group Patients overestimated the
(608) groups; standardized Adult patients with findings and the results of benefits and felt uninformed
23420455 patients (providers) ICDs standardized patient interviews regarding the risks.
Cardiologists Patient-physician
Aim: To examine Results - Patients: communication about ICDs is
patient-physician Exclusion criteria: N/A 33/41 patients could not recall a characterized by unclear
communication at the discussion about complications. representation and omission of
time the decision is Patients felt a score of 5.7 on a scale information to patients
made to implant an of 1-10 on feeling informed
ICD. Mean number of patients out of 100
who would be saved by the ICD was
Size: 41 patients, 11 87.9
providers
Results - Clinicians:
In 17 of 22 of interviews,
cardiologists did not address or
minimized or denied QOL issues
and long-term consequences of
ICD placement
In 15 of 22 of the standardized
patient interviews, cardiologists

315

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
used unexplained medical terms
or jargon.
Stewart et al. 2010 Study type: Survey Inclusion criteria: 1 endpoint/Results Study demonstrated that
(609) Patients with EF Most patients anticipated more than patients overestimate the
20142021 Aim: To examine <35% 10 y survival. benefits of ICD therapy.
patient expectations Symptomatic HF 54% expected an ICD to save 50 lives
from ICDs for 1 per 100 during 5 y.
prevention of sudden Exclusion criteria: N/A 70% of ICD recipients indicated they
death in HF. would keep the ICD on even if dying of
cancer,
Size: 105 55% even if having daily shocks,
None would inactivate even if
suffering constant dyspnea at rest.
Ottenberg et al. 2014 Study type: Inclusion criteria: 1 endpoint/Results: 5 Themes: Interviews identified significant
(610) Qualitative Focus Patients who had (1) don't mess with a good thing; gaps for some patients in their
24889010 Group declined ICD (12 ICD, one (2) my health is good enough; understanding about the ICD.
CRT) (3) independent decision making;
Aim: To describe the (4) it's your job, but it's my choice;
reasons why patients Exclusion criteria: N/A and
decline ICD (5) gaps in learning
implantation

Size: 13 patients (3
groups)
Yuhas et al. 2012 Study type: Inclusion criteria: 1 Endpoint/Results: 5 Themes: People who decline had
(611) Qualitative interview outpatient cardiology (1) Patients who refused ICD referral misunderstandings about their
22897624 patients with EF 35% had a lack of insight into their own personal risk.
Aim: To explore and without an ICD. risk.
patients attitudes and (2) Many patients who accepted ICD
perceptions of ICDs to Exclusion criteria: N/A referral perceived that this was
better understand strongly recommended by their
potential patient- physicians.
related barriers to (3) Concerns over recall, malfunction,
appropriate utilization. and surgical risk were common in
both.
Size: N=25. 12 who
accepted referral, 13

316

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
who declined referral (4) Many patients demonstrated
(note: none had ICDs) inaccurate perceptions of ICD-related
risks
(5) Feelings regarding invasive life-
prolonging interventions played an
important role in ICD referral refusal
for some individuals.

Data Supplement 61. Randomized Trials, Observational Studies, and/or Registries Related to Cost and Value Considerations - (Section 16)
Study Study Design Patient Population Costs Effectiveness Value Summary/Conclusions
Name Study Size
AVID Study type: RCT of ICD vs. 2 prevention: Within trial: Within trial: Lifetime ICER= Intermediate value
Larsen G, et al. antiarrhythimic drug resuscitated CA or ICD $87,479, ICD 2.48 y, $67,100 based on ACC/AHA
2002 (612) therapy (largely sustained VT, EF 40%. Antiarrythmic Antiarrythmic benchmarks.
11980684 amiodarone). drug Tx drug Tx 2.27 y Within-trial Authors concluded:
$73,564 ICER= $66,700 ICD was moderately
Within trial costs and cost-effective for 2
outcomes to 3 y; lifetime prevention.
projection.

Size:
1,008 patients

CIDS Study type: 2 prevention: Within trial: Within trial: 12 year ICER; Intermediate value
OBrien BJ, et RCT of ICD vs. Resuscitated VF or VT. ICD C$87,715; ICD 4.58 y; C$99,400 based on ACC/AHA
al. 2001 (613) amiodarone. amiodarone amiodarone (US$67,600) benchmarks.
11245646 C$38,600 4.35 y (with continued Authors concluded
Within trial cost and ICD benefit) that ICD therapy is not
survival to 6 y; 12 y attractive based on
projection of cost and Within trial Canadian standards.
survival. ICER= No lifetime
430 patients in economic C$213,500 projections of cost and
substudy. (US$145,200) life expectancy.
Size: 659 total patients

317

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Weiss, et al. Study type: 2 prevention. Within study: Within study: Within study Intermediate value
2002 (614) Propensity score matched Hospitalized with 1 ICD $78,700; ICD 4.6 y; ICER= $78,400 based on ACC/AHA
12015242 analysis of Medicare diagnosis of VT or VF. conventional conventional benchmarks.
patients. Costs and therapy therapy 4.1 y No lifetime
outcomes to 8 y. $37,200 projections of cost and
life expectancy.
Size: 7,619 matched pairs
Buxton et al. Study type: Markov 2 prevention. ICD: 87,184; Life-y: ICD 9.87; 48,700/life-y Intermediate value
2006 (615) model, 20 y time amiodarone: amiodarone gained based on ACC/AHA
16904046 horizons. Effectiveness 18,379 8.41 ($64,700) benchmarks.
inputs from RCTs, cost Authors concluded
inputs from UK. Quality- 65,000/QALY that ICDs were not
adjusted life-y: gained cost-effective at the UK
Size: Cost data from 535 ICD 7.41, ($86,200) benchmark (<30,000).
patients with ICD amiodarone
implants in Liverpool. 6.35
SCD-HeFT Study type: 1 prevention: HF Within trial: Life expectancy: Lifetime ICER= High value based on
Mark DB, et al. RCT of ICD vs. (NYHA II or III) and EF ICD $61,938; ICD 10.87 y; $38,400 ACC/AHA benchmarks.
(616) amiodarone or placebo. 35%. placebo placebo 8.41 y Authors concluded
16818817 $42,971 Within trial that ICD was
Costs and outcomes to 5 ICER= $127,500 economically
y; lifetime projection of Lifetime: attractive compared
costs and life expectancy. ICD $158,840; with placebo as long as
1,692 patients in placebo ICD benefit was
economic substudy (US $79,028 maintained for 8 y.
centers),

Size: 2,521 total patients


MADIT-II Study type: RCT of ICD vs 1 prevention: Patients Within trial: Within trial: 12 y ICER= Intermediate value
Zwanziger J, et conventional medical with prior MI, EF 30%. ICD $84,100, ICD 2.89 y, $78,600 to based on ACC/AHA
al. 2006 (617) therapy. conventional conventional $114,000 benchmarks, based on
16750701 $44,900; 2.72 y long-term projections
Within trial costs and Within trial of ICD outcomes.
survival to 3.5 y; 12 y 12 year ICER
projection of cost and projections: =$235,000;
survival. ICD $173,700
to $180,300,
318

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
Size: 1,095 patients in conventional
economic substudy (US $97,900
patients),
1,232 total patients
MADIT-I Study type: 1 prevention. Prior MI, Within trial: Within trial: Within trial High value based on
Mushlin AI, et RCT of ICD or medical asymptomatic non- ICD $97,560; ICD 3.66 y, ICER= ACC/AHA benchmarks.
al. 1998 (618) therapy. sustained VT, EF 35%, medical medical therapy $27,000 Authors concluded
9626173 inducible VT not therapy 2.80 y that ICD is cost-
Costs and outcomes to 4 suppressed by $78,980 effective in selected
y. procainamide. individuals at high risk
for sudden cardiac
Size: death.
181 patients in economic
study (US centers), 196
total patients.
Al-Khatib, et Study type: 1 prevention. Post-MI, ICD: Life expectancy: $50,500 per Intermediate value
al. 2005 (619) Duke database outcomes EF 30%. $131,490; ICD 8.59 y, life-y gained by ACC/AHA
15838065 and costs for 15 y. medical: medical 6.79 y benchmarks
Llifetime extrapolation by $40,661 Authors concluded:
Markov model. ICD therapy for patients
eligible for MADIT-II
Size: 1,285 patients was economically
attractive by
conventional
standards.
Sanders, et al. Study type: 1 prevention. Trial ICD had higher ICD had higher $39,000 for High value by
2005 (620) Markov model, lifetime subjects in CABG-PATCH, costs in each life expectancy COMPANION, ACC/AHA benchmarks
16207849 projection, applied to COMPANION, DEFINITE, population: in six trials, DEFINITE, when projected life
data from each of eight DINAMIT, MADIT-I, $55,700 to ranging from MADIT I, MADIT expectancy was
randomized trials. MADIT-II, MUSTT, and $100,500 1.46 to 4.14 life- II, MUSTT; increased by >1.4 y
SCD-HeFT. y added
Size: Not applicable $50,700 for
SCD-HeFT

Higher cost,
worse
outcomes for

319

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
CABG-PATCH,
DINAMIT.
Smith, et al. Study type: 1 prevention. Patients ICD 86,759; ICD 7.08 QALY; ICER= 44,000 High value by
2013 (621) Markov model, lifetime with EF <40%, due to conventional conventional ($49,200) ACC/AHA benchmarks.
22584647 projection. Effectiveness either ischemic or non- therapy therapy 6.26 Authors concluded:
from meta-analysis of 6 ischemic causes. 50,685 QALY 1 prophylactic ICD
RCTs. therapy had high value
in the European setting
Size: Not applicable for patients with EF
<40%.
Cowie, et Study type: 1 prevention. Patients ICD 64,600; ICD 8.58 life-y ICER= 24,800/ High value by
al.2009 (622) Markov model, lifetime with EF <35%, ischemic conventional (7.27 QALY); life-y gained ACC/AHA benchmarks.
19359333 projection. Effectiveness or non-ischemic therapy conventional ($27,700) Authors concluded:
from meta-analysis of 6 etiology. 18,187 therapy 6.71 Prophylactic ICD
RCTs. European costs. life-y 29,500/QALY implantation had high
(5.70 QALY) gained value if current
Size: Not applicable ($33,000) guidelines for patients
with EF <35% are
followed.

320

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
References:
1. Ruwald MH, Hansen ML, Lamberts M, et al. The relation between age, sex, comorbidity, and pharmacotherapy and the risk of syncope: a
Danish nationwide study. Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on
cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2012; 14:1506-14.
2. Soteriades ES, Evans JC, Larson MG, et al. Incidence and prognosis of syncope. N. Engl. J. Med. 2002; 347:878-85.
3. Middlekauff HR, Stevenson WG, Stevenson LW, et al. Syncope in advanced heart failure: high risk of sudden death regardless of origin of
syncope. J Am Coll Cardiol. 1993; 21:110-6.
4. Steinman RT, Herrera C, Schuger CD, et al. Wide QRS tachycardia in the conscious adult. Ventricular tachycardia is the most frequent
cause. Jama. 1989; 261:1013-6.
5. Brugada P, Brugada J, Mont L, et al. A new approach to the differential diagnosis of a regular tachycardia with a wide QRS complex.
Circulation. 1991; 83:1649-59.
6. Wellens HJ, Bar FW and Lie KI. The value of the electrocardiogram in the differential diagnosis of a tachycardia with a widened QRS
complex. Am J Med. 1978; 64:27-33.
7. Elhendy A, Chandrasekaran K, Gersh BJ, et al. Functional and prognostic significance of exercise-induced ventricular arrhythmias in
patients with suspected coronary artery disease. Am. J. Cardiol. 2002; 90:95-100.
8. Grady TA, Chiu AC, Snader CE, et al. Prognostic significance of exercise-induced left bundle-branch block. Jama. 1998; 279:153-6.
9. Costantini O, Hohnloser SH, Kirk MM, et al. The ABCD (Alternans Before Cardioverter Defibrillator) Trial: strategies using T-wave alternans
to improve efficiency of sudden cardiac death prevention. J Am Coll Cardiol. 2009; 53:471-9.
10. Desai AD, Yaw TS, Yamazaki T, et al. Prognostic significance of quantitative QRS duration. Am J Med. 2006; 119:600-6.
11. Freedman RA, Alderman EL, Sheffield LT, et al. Bundle branch block in patients with chronic coronary artery disease: angiographic
correlates and prognostic significance. J Am Coll Cardiol. 1987; 10:73-80.
12. Baldasseroni S, Opasich C, Gorini M, et al. Left bundle-branch block is associated with increased 1-year sudden and total mortality rate in
5517 outpatients with congestive heart failure: a report from the Italian network on congestive heart failure. Am. Heart J. 2002; 143:398-
405.
13. Zimetbaum PJ, Buxton AE, Batsford W, et al. Electrocardiographic predictors of arrhythmic death and total mortality in the multicenter
unsustained tachycardia trial. Circulation. 2004; 110:766-9.
14. Buxton AE, Sweeney MO, Wathen MS, et al. QRS duration does not predict occurrence of ventricular tachyarrhythmias in patients with
implanted cardioverter-defibrillators. J Am Coll Cardiol. 2005; 46:310-6.
15. Monasterio V, Martinez JP, Laguna P, et al. Prognostic value of average T-wave alternans and QT variability for cardiac events in MADIT-II
patients. Journal of electrocardiology. 2013; 46:480-6.
16. Chow T, Kereiakes DJ, Onufer J, et al. Does microvolt T-wave alternans testing predict ventricular tachyarrhythmias in patients with
ischemic cardiomyopathy and prophylactic defibrillators? The MASTER (Microvolt T Wave Alternans Testing for Risk Stratification of Post-
Myocardial Infarction Patients) trial. J Am Coll Cardiol. 2008; 52:1607-15.
321

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
17. Gupta A, Hoang DD, Karliner L, et al. Ability of microvolt T-wave alternans to modify risk assessment of ventricular tachyarrhythmic events:
a meta-analysis. Am. Heart J. 2012; 163:354-64.
18. Dhar R, Alsheikh-Ali AA, Estes NA, III, et al. Association of prolonged QRS duration with ventricular tachyarrhythmias and sudden cardiac
death in the Multicenter Automatic Defibrillator Implantation Trial II (MADIT-II). Heart rhythm. 2008; 5:807-13.
19. Bloomfield DM, Steinman RC, Namerow PB, et al. Microvolt T-wave alternans distinguishes between patients likely and patients not likely
to benefit from implanted cardiac defibrillator therapy: a solution to the Multicenter Automatic Defibrillator Implantation Trial (MADIT) II
conundrum. Circulation. 2004; 110:1885-9.
20. Iuliano S, Fisher SG, Karasik PE, et al. QRS duration and mortality in patients with congestive heart failure. Am. Heart J. 2002; 143:1085-91.
21. Perez-Rodon J, Martinez-Alday J, Baron-Esquivias G, et al. Prognostic value of the electrocardiogram in patients with syncope: data from
the group for syncope study in the emergency room (GESINUR). Heart rhythm. 2014; 11:2035-44.
22. Barrett PM, Komatireddy R, Haaser S, et al. Comparison of 24-hour Holter monitoring with 14-day novel adhesive patch
electrocardiographic monitoring. Am J Med. 2014; 127:95-7.
23. de Asmundis C, Conte G, Sieira J, et al. Comparison of the patient-activated event recording system vs. traditional 24 h Holter
electrocardiography in individuals with paroxysmal palpitations or dizziness. Europace : European pacing, arrhythmias, and cardiac
electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European
Society of Cardiology. 2014; 16:1231-5.
24. Turakhia MP, Hoang DD, Zimetbaum P, et al. Diagnostic utility of a novel leadless arrhythmia monitoring device. Am. J. Cardiol. 2013;
112:520-4.
25. Linzer M, Pritchett EL, Pontinen M, et al. Incremental diagnostic yield of loop electrocardiographic recorders in unexplained syncope. Am.
J. Cardiol. 1990; 66:214-9.
26. Bloch Thomsen PE, Jons C, Raatikainen MJ, et al. Long-term recording of cardiac arrhythmias with an implantable cardiac monitor in
patients with reduced ejection fraction after acute myocardial infarction: the Cardiac Arrhythmias and Risk Stratification After Acute
Myocardial Infarction (CARISMA) study. Circulation. 2010; 122:1258-64.
27. Volosin K, Stadler RW, Wyszynski R, et al. Tachycardia detection performance of implantable loop recorders: results from a large 'real-life'
patient cohort and patients with induced ventricular arrhythmias. Europace : European pacing, arrhythmias, and cardiac
electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European
Society of Cardiology. 2013; 15:1215-22.
28. Krahn AD, Klein GJ, Yee R, et al. Use of an extended monitoring strategy in patients with problematic syncope. Reveal Investigators.
Circulation. 1999; 99:406-10.
29. Solbiati M, Costantino G, Casazza G, et al. Implantable loop recorder versus conventional diagnostic workup for unexplained recurrent
syncope. Cochrane Database Syst. Rev. 2016; 4:CD011637.
30. Solomon SD, Zelenkofske S, McMurray JJ, et al. Sudden death in patients with myocardial infarction and left ventricular dysfunction, heart
failure, or both. N. Engl. J. Med. 2005; 352:2581-8.

322

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
31. Gula LJ, Klein GJ, Hellkamp AS, et al. Ejection fraction assessment and survival: an analysis of the Sudden Cardiac Death in Heart Failure
Trial (SCD-HeFT). Am. Heart J. 2008; 156:1196-200.
32. Korngold EC, Januzzi JL, Jr., Gantzer ML, et al. Amino-terminal pro-B-type natriuretic peptide and high-sensitivity C-reactive protein as
predictors of sudden cardiac death among women. Circulation. 2009; 119:2868-76.
33. Patton KK, Sotoodehnia N, DeFilippi C, et al. N-terminal pro-B-type natriuretic peptide is associated with sudden cardiac death risk: the
Cardiovascular Health Study. Heart rhythm. 2011; 8:228-33.
34. Scott PA, Barry J, Roberts PR, et al. Brain natriuretic peptide for the prediction of sudden cardiac death and ventricular arrhythmias: a
meta-analysis. Eur. J Heart Fail. 2009; 11:958-66.
35. Blangy H, Sadoul N, Dousset B, et al. Serum BNP, hs-C-reactive protein, procollagen to assess the risk of ventricular tachycardia in ICD
recipients after myocardial infarction. Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working
groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2007; 9:724-9.
36. Ahmad T, Fiuzat M, Neely B, et al. Biomarkers of myocardial stress and fibrosis as predictors of mode of death in patients with chronic
heart failure. JACC. Heart Fail. 2014; 2:260-8.
37. Levine YC, Rosenberg MA, Mittleman M, et al. B-type natriuretic peptide is a major predictor of ventricular tachyarrhythmias. Heart
rhythm. 2014; 11:1109-16.
38. Berger R, Huelsman M, Strecker K, et al. B-type natriuretic peptide predicts sudden death in patients with chronic heart failure. Circulation.
2002; 105:2392-7.
39. Buxton AE, Lee KL, Hafley GE, et al. Relation of ejection fraction and inducible ventricular tachycardia to mode of death in patients with
coronary artery disease: an analysis of patients enrolled in the multicenter unsustained tachycardia trial. Circulation. 2002; 106:2466-72.
40. Buxton AE, Lee KL, Fisher JD, et al. A randomized study of the prevention of sudden death in patients with coronary artery disease.
Multicenter Unsustained Tachycardia Trial Investigators. N. Engl. J. Med. 1999; 341:1882-90.
41. Buxton AE, Lee KL, DiCarlo L, et al. Electrophysiologic testing to identify patients with coronary artery disease who are at risk for sudden
death. Multicenter Unsustained Tachycardia Trial Investigators. N. Engl. J. Med. 2000; 342:1937-45.
42. Moss AJ, Hall WJ, Cannom DS, et al. Improved survival with an implanted defibrillator in patients with coronary disease at high risk for
ventricular arrhythmia. Multicenter Automatic Defibrillator Implantation Trial Investigators. N. Engl. J. Med. 1996; 335:1933-40.
43. Bardy GH, Lee KL, Mark DB, et al. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N. Engl. J. Med.
2005; 352:225-37.
44. Moss AJ, Zareba W, Hall WJ, et al. Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection
fraction. N. Engl. J. Med. 2002; 346:877-83.
45. Hilfiker G, Schoenenberger AW, Erne P, et al. Utility of electrophysiological studies to predict arrhythmic events. World J Cardiol. 2015;
7:344-50.
46. Bourke JP, Richards DA, Ross DL, et al. Routine programmed electrical stimulation in survivors of acute myocardial infarction for prediction
of spontaneous ventricular tachyarrhythmias during follow-up: results, optimal stimulation protocol and cost-effective screening. J Am Coll
Cardiol. 1991; 18:780-8.
323

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
47. Bailey JJ, Berson AS, Handelsman H, et al. Utility of current risk stratification tests for predicting major arrhythmic events after myocardial
infarction. J Am Coll Cardiol. 2001; 38:1902-11.
48. Schmitt C, Barthel P, Ndrepepa G, et al. Value of programmed ventricular stimulation for prophylactic internal cardioverter-defibrillator
implantation in postinfarction patients preselected by noninvasive risk stratifiers. J Am Coll Cardiol. 2001; 37:1901-7.
49. Brembilla-Perrot B, Suty-Selton C, Beurrier D, et al. Differences in mechanisms and outcomes of syncope in patients with coronary disease
or idiopathic left ventricular dysfunction as assessed by electrophysiologic testing. J Am Coll Cardiol. 2004; 44:594-601.
50. Bhandari AK, Shapiro WA, Morady F, et al. Electrophysiologic testing in patients with the long QT syndrome. Circulation. 1985; 71:63-71.
51. Giustetto C, Di MF, Wolpert C, et al. Short QT syndrome: clinical findings and diagnostic-therapeutic implications. Eur. Heart J. 2006;
27:2440-7.
52. Mahida S, Derval N, Sacher F, et al. Role of electrophysiological studies in predicting risk of ventricular arrhythmia in early repolarization
syndrome. J Am Coll Cardiol. 2015; 65:151-9.
53. Giustetto C, Schimpf R, Mazzanti A, et al. Long-term follow-up of patients with short QT syndrome. J Am Coll Cardiol. 2011; 58:587-95.
54. Raczak G, Pinna GD, Maestri R, et al. Different predictive values of electrophysiological testing and autonomic assessment in patients
surviving a sustained arrhythmic episode. Circ. J. 2004; 68:634-8.
55. Brodsky MA, Mitchell LB, Halperin BD, et al. Prognostic value of baseline electrophysiology studies in patients with sustained ventricular
tachyarrhythmia: the Antiarrhythmics Versus Implantable Defibrillators (AVID) trial. Am. Heart J. 2002; 144:478-84.
56. Daubert JP, Zareba W, Hall WJ, et al. Predictive value of ventricular arrhythmia inducibility for subsequent ventricular tachycardia or
ventricular fibrillation in Multicenter Automatic Defibrillator Implantation Trial (MADIT) II patients. J Am Coll Cardiol. 2006; 47:98-107.
57. Daubert JP, Winters SL, Subacius H, et al. Ventricular arrhythmia inducibility predicts subsequent ICD activation in nonischemic
cardiomyopathy patients: a DEFINITE substudy. Pacing Clin. Electrophysiol. 2009; 32:755-61.
58. Gold MR, Bloomfield DM, Anderson KP, et al. A comparison of T-wave alternans, signal averaged electrocardiography and programmed
ventricular stimulation for arrhythmia risk stratification. J Am Coll Cardiol. 2000; 36:2247-53.
59. Gatzoulis KA, Vouliotis AI, Tsiachris D, et al. Primary prevention of sudden cardiac death in a nonischemic dilated cardiomyopathy
population: reappraisal of the role of programmed ventricular stimulation. Circ. Arrhythm. Electrophysiol. 2013; 6:504-12.
60. Dargie HJ. Effect of carvedilol on outcome after myocardial infarction in patients with left-ventricular dysfunction: the CAPRICORN
randomised trial. Lancet (London, England). 2001; 357:1385-90.
61. Packer M, Bristow MR, Cohn JN, et al. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S.
Carvedilol Heart Failure Study Group. N. Engl. J. Med. 1996; 334:1349-55.
62. The cardiac insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet (London, England). 1999; 353:9-13.
63. Hjalmarson A, Goldstein S, Fagerberg B, et al. Effects of controlled-release metoprolol on total mortality, hospitalizations, and well-being in
patients with heart failure: the Metoprolol CR/XL Randomized Intervention Trial in congestive heart failure (MERIT-HF). MERIT-HF Study
Group. Jama. 2000; 283:1295-302.
64. Cohn JN, Johnson G, Ziesche S, et al. A comparison of enalapril with hydralazine-isosorbide dinitrate in the treatment of chronic congestive
heart failure. N. Engl. J. Med. 1991; 325:303-10.
324

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
65. Cohn JN and Tognoni G. A randomized trial of the angiotensin-receptor blocker valsartan in chronic heart failure. N. Engl. J. Med. 2001;
345:1667-75.
66. Pfeffer MA, McMurray JJ, Velazquez EJ, et al. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left
ventricular dysfunction, or both. N. Engl. J. Med. 2003; 349:1893-906.
67. Pitt B, Segal R, Martinez FA, et al. Randomised trial of losartan versus captopril in patients over 65 with heart failure (Evaluation of
Losartan in the Elderly Study, ELITE). Lancet (London, England). 1997; 349:747-52.
68. Pitt B, Poole-Wilson PA, Segal R, et al. Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure:
randomised trial--the Losartan Heart Failure Survival Study ELITE II. Lancet (London, England). 2000; 355:1582-7.
69. Pitt B, Zannad F, Remme WJ, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure.
Randomized Aldactone Evaluation Study Investigators. N. Engl. J. Med. 1999; 341:709-17.
70. Pitt B, Remme W, Zannad F, et al. Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial
infarction. N. Engl. J. Med. 2003; 348:1309-21.
71. Zannad F, McMurray JJ, Krum H, et al. Eplerenone in patients with systolic heart failure and mild symptoms. N. Engl. J. Med. 2011; 364:11-
21.
72. Desai AS, McMurray JJ, Packer M, et al. Effect of the angiotensin-receptor-neprilysin inhibitor LCZ696 compared with enalapril on mode of
death in heart failure patients. Eur. Heart J. 2015; 36:1990-7.
73. Carson P, Wertheimer J, Miller A, et al. The STICH trial (Surgical Treatment for Ischemic Heart Failure): mode-of-death results. JACC. Heart
Fail. 2013; 1:400-8.
74. Velazquez EJ, Lee KL, Jones RH, et al. Coronary-artery bypass surgery in patients with ischemic cardiomyopathy. N. Engl. J. Med. 2016;
374:1511-20.
75. Cook JR, Rizo-Patron C, Curtis AB, et al. Effect of surgical revascularization in patients with coronary artery disease and ventricular
tachycardia or fibrillation in the Antiarrhythmics Versus Implantable Defibrillators (AVID) Registry. Am. Heart J. 2002; 143:821-6.
76. Mondsert B, Khairy P, Schram G, et al. Impact of revascularization in patients with sustained ventricular arrhythmias, prior myocardial
infarction, and preserved left ventricular ejection fraction. Heart rhythm. 2016; 13:1221-7.
77. Ngaage DL, Cale AR, Cowen ME, et al. Early and late survival after surgical revascularization for ischemic ventricular fibrillation/tachycardia.
Ann. Thorac. Surg. 2008; 85:1278-81.
78. Every NR, Fahrenbruch CE, Hallstrom AP, et al. Influence of coronary bypass surgery on subsequent outcome of patients resuscitated from
out of hospital cardiac arrest. J Am Coll Cardiol. 1992; 19:1435-9.
79. van der Burg AE, Bax JJ, Boersma E, et al. Impact of viability, ischemia, scar tissue, and revascularization on outcome after aborted sudden
death. Circulation. 2003; 108:1954-9.
80. Dumas F, Cariou A, Manzo-Silberman S, et al. Immediate percutaneous coronary intervention is associated with better survival after out-
of-hospital cardiac arrest: insights from the PROCAT (Parisian Region Out of hospital Cardiac ArresT) registry. Circ. Cardiovasc Interv. 2010;
3:200-7.

325

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
81. Dumas F, Bougouin W, Geri G, et al. Emergency PCI in post-cardiac arrest patients without ST-segment elevation pattern: insights from the
PROCAT II registry. JACC. Cardiovasc. Interv. 2016; 9:1011-8.
82. Serruys PW, Morice MC, Kappetein AP, et al. Percutaneous coronary intervention versus coronary-artery bypass grafting for severe
coronary artery disease. N. Engl. J. Med. 2009; 360:961-72.
83. Milojevic M, Head SJ, Parasca CA, et al. Causes of death following PCI versus CABG in complex CAD: 5-year follow-up of SYNTAX. J Am Coll
Cardiol. 2016; 67:42-55.
84. Al-Khatib SM, Hellkamp AS, Lee KL, et al. Implantable cardioverter defibrillator therapy in patients with prior coronary revascularization in
the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT). J. Cardiovasc. Electrophysiol. 2008; 19:1059-65.
85. Nageh MF, Kim JJ, Chen LH, et al. Implantable defibrillators for secondary prevention of sudden cardiac death in cardiac surgery patients
with perioperative ventricular arrhythmias. Journal of the American Heart Association. 2014; 3.
86. Kumar S, Barbhaiya CR, Sobieszczyk P, et al. Role of alternative interventional procedures when endo- and epicardial catheter ablation
attempts for ventricular arrhythmias fail. Circ. Arrhythm. Electrophysiol. 2015; 8:606-15.
87. Anter E, Hutchinson MD, Deo R, et al. Surgical ablation of refractory ventricular tachycardia in patients with nonischemic cardiomyopathy.
Circ. Arrhythm. Electrophysiol. 2011; 4:494-500.
88. Bhavani SS, Tchou P, Saliba W, et al. Surgical options for refractory ventricular tachycardia. J Card Surg. 2007; 22:533-4.
89. Sartipy U, Albage A, Straat E, et al. Surgery for ventricular tachycardia in patients undergoing left ventricular reconstruction by the Dor
procedure. Ann. Thorac. Surg. 2006; 81:65-71.
90. Choi EK, Nagashima K, Lin KY, et al. Surgical cryoablation for ventricular tachyarrhythmia arising from the left ventricular outflow tract
region. Heart rhythm. 2015; 12:1128-36.
91. Patel M, Rojas F, Shabari FR, et al. Safety and Feasibility of Open Chest Epicardial Mapping and Ablation of Ventricular Tachycardia During
the Period of Left Ventricular Assist Device Implantation. J. Cardiovasc. Electrophysiol. 2016; 27:95-101.
92. Mulloy DP, Bhamidipati CM, Stone ML, et al. Cryoablation during left ventricular assist device implantation reduces postoperative
ventricular tachyarrhythmias. J Thorac. Cardiovasc. Surg. 2013; 145:1207-13.
93. Schwartz PJ, Motolese M and Pollavini G. Prevention of Sudden Cardiac Death After a First Myocardial Infarction by Pharmacologic or
Surgical Antiadrenergic Interventions. Journal of cardiovascular electrophysiology. 1992; 3:2-16.
94. Krittayaphong R, Bhuripanyo K, Punlee K, et al. Effect of atenolol on symptomatic ventricular arrhythmia without structural heart disease:
a randomized placebo-controlled study. Am. Heart J. 2002; 144:e10.
95. Vaseghi M, Gima J, Kanaan C, et al. Cardiac sympathetic denervation in patients with refractory ventricular arrhythmias or electrical storm:
intermediate and long-term follow-up. Heart rhythm. 2014; 11:360-6.
96. Ajijola OA, Lellouche N, Bourke T, et al. Bilateral cardiac sympathetic denervation for the management of electrical storm. J Am Coll
Cardiol. 2012; 59:91-2.
97. Ukena C, Mahfoud F, Ewen S, et al. Renal denervation for treatment of ventricular arrhythmias: data from an International Multicenter
Registry. Clin. Res. Cardiol. 2016.
98. Grimaldi R, de LA, Kornet L, et al. Can spinal cord stimulation reduce ventricular arrhythmias? Heart rhythm. 2012; 9:1884-7.
326

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
99. Kudenchuk PJ, Brown SP, Daya M, et al. Amiodarone, lidocaine, or placebo in out-of-hospital cardiac arrest. N. Engl. J. Med. 2016:1711-22.
100. Joglar JA and Page RL. Out-of-Hospital Cardiac Arrest--Are Drugs Ever the Answer? N. Engl. J. Med. 2016; 374:1781-2.
101. Jacobs IG, Finn JC, Jelinek GA, et al. Effect of adrenaline on survival in out-of-hospital cardiac arrest: A randomised double-blind placebo-
controlled trial. Resuscitation. 2011; 82:1138-43.
102. Piccini JP, Hranitzky PM, Kilaru R, et al. Relation of mortality to failure to prescribe beta blockers acutely in patients with sustained
ventricular tachycardia and ventricular fibrillation following acute myocardial infarction (from the VALsartan In Acute myocardial
iNfarcTion trial [VALIANT] Registry). Am J Cardiol. 2008; 102:1427-32.
103. Dorian P, Cass D, Schwartz B, et al. Amiodarone as compared with lidocaine for shock-resistant ventricular fibrillation. N. Engl. J. Med.
2002; 346:884-90.
104. Hassan TB, Jagger C and Barnett DB. A randomised trial to investigate the efficacy of magnesium sulphate for refractory ventricular
fibrillation. Emerg. Med. J. 2002; 19:57-62.
105. Thel MC, Armstrong AL, McNulty SE, et al. Randomised trial of magnesium in in-hospital cardiac arrest. Duke Internal Medicine Housestaff.
Lancet (London, England). 1997; 350:1272-6.
106. Somberg JC, Bailin SJ, Haffajee CI, et al. Intravenous lidocaine versus intravenous amiodarone (in a new aqueous formulation) for incessant
ventricular tachycardia. Am. J. Cardiol. 2002; 90:853-9.
107. Kudenchuk PJ, Cobb LA, Copass MK, et al. Amiodarone for resuscitation after out-of-hospital cardiac arrest due to ventricular fibrillation.
N. Engl. J. Med. 1999; 341:871-8.
108. Callaham M, Madsen CD, Barton CW, et al. A randomized clinical trial of high-dose epinephrine and norepinephrine vs standard-dose
epinephrine in prehospital cardiac arrest. Jama. 1992; 268:2667-72.
109. Gueugniaud PY, Mols P, Goldstein P, et al. A comparison of repeated high doses and repeated standard doses of epinephrine for cardiac
arrest outside the hospital. European Epinephrine Study Group. N. Engl. J. Med. 1998; 339:1595-601.
110. Gorgels AP, van den Dool A, Hofs A, et al. Comparison of procainamide and lidocaine in terminating sustained monomorphic ventricular
tachycardia. Am. J. Cardiol. 1996; 78:43-6.
111. Ho DS, Zecchin RP, Richards DA, et al. Double-blind trial of lignocaine versus sotalol for acute termination of spontaneous sustained
ventricular tachycardia. Lancet (London, England). 1994; 344:18-23.
112. Levine JH, Massumi A, Scheinman MM, et al. Intravenous amiodarone for recurrent sustained hypotensive ventricular tachyarrhythmias.
Intravenous Amiodarone Multicenter Trial Group. J Am Coll Cardiol. 1996; 27:67-75.
113. Teo KK, Yusuf S and Furberg CD. Effects of prophylactic antiarrhythmic drug therapy in acute myocardial infarction. An overview of results
from randomized controlled trials. Jama. 1993; 270:1589-95.
114. Elizari MV, Martinez JM, Belziti C, et al. Morbidity and mortality following early administration of amiodarone in acute myocardial
infarction. GEMICA study investigators, GEMA Group, Buenos Aires, Argentina. Grupo de Estudios Multicentricos en Argentina. Eur. Heart
J. 2000; 21:198-205.
115. Link MS, Berkow LC, Kudenchuk PJ, et al. Part 7: Adult Advanced Cardiovascular Life Support: 2015 American Heart Association Guidelines
Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation. 2015; 132:S444-S64.
327

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
116. Herlitz J, Ekstrom L, Wennerblom B, et al. Lidocaine in out-of-hospital ventricular fibrillation. Does it improve survival? Resuscitation. 1997;
33:199-205.
117. Markel DT, Gold LS, Allen J, et al. Procainamide and survival in ventricular fibrillation out-of-hospital cardiac arrest. Acad. Emerg. Med.
2010; 17:617-23.
118. Stiell IG, Wells GA, Field B, et al. Advanced cardiac life support in out-of-hospital cardiac arrest. N. Engl. J. Med. 2004; 351:647-56.
119. Hagihara A, Hasegawa M, Abe T, et al. Prehospital epinephrine use and survival among patients with out-of-hospital cardiac arrest. Jama.
2012; 307:1161-8.
120. Donnino MW, Salciccioli JD, Howell MD, et al. Time to administration of epinephrine and outcome after in-hospital cardiac arrest with non-
shockable rhythms: retrospective analysis of large in-hospital data registry. BMJ. 2014; 348:g3028.
121. Koscik C, Pinawin A, McGovern H, et al. Rapid epinephrine administration improves early outcomes in out-of-hospital cardiac arrest.
Resuscitation. 2013; 84:915-20.
122. Spaulding CM, Joly LM, Rosenberg A, et al. Immediate coronary angiography in survivors of out-of-hospital cardiac arrest. N. Engl. J. Med.
1997; 336:1629-33.
123. Cronier P, Vignon P, Bouferrache K, et al. Impact of routine percutaneous coronary intervention after out-of-hospital cardiac arrest due to
ventricular fibrillation. Crit. Care. 2011; 15:R122.
124. Zanuttini D, Armellini I, Nucifora G, et al. Impact of emergency coronary angiography on in-hospital outcome of unconscious survivors after
out-of-hospital cardiac arrest. Am. J. Cardiol. 2012; 110:1723-8.
125. Kudenchuk PJ, Newell C, White L, et al. Prophylactic lidocaine for post resuscitation care of patients with out-of-hospital ventricular
fibrillation cardiac arrest. Resuscitation. 2013; 84:1512-8.
126. Nademanee K, Taylor R, Bailey WE, et al. Treating electrical storm : sympathetic blockade versus advanced cardiac life support-guided
therapy. Circulation. 2000; 102:742-7.
127. Sasson C, Rogers MA, Dahl J, et al. Predictors of survival from out-of-hospital cardiac arrest: a systematic review and meta-analysis. Circ.
Cardiovasc. Qual. Outcomes. 2010; 3:63-81.
128. Buxton AE, Marchlinski FE, Doherty JU, et al. Hazards of intravenous verapamil for sustained ventricular tachycardia. Am. J. Cardiol. 1987;
59:1107-10.
129. Pellis T, Kette F, Lovisa D, et al. Utility of pre-cordial thump for treatment of out of hospital cardiac arrest: a prospective study.
Resuscitation. 2009; 80:17-23.
130. Volkmann H, Klumbies A, Kuhnert H, et al. Terminating ventricular tachycardias by mechanical heart stimulation with precordial thumps. Z.
Kardiol. 1990; 79:717-24.
131. A comparison of antiarrhythmic-drug therapy with implantable defibrillators in patients resuscitated from near-fatal ventricular
arrhythmias. The Antiarrhythmics versus Implantable Defibrillators (AVID) Investigators. N. Engl. J. Med. 1997; 337:1576-83.
132. Connolly SJ, Gent M, Roberts RS, et al. Canadian implantable defibrillator study (CIDS) : a randomized trial of the implantable cardioverter
defibrillator against amiodarone. Circulation. 2000; 101:1297-302.

328

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
133. Kuck KH, Cappato R, Siebels J, et al. Randomized comparison of antiarrhythmic drug therapy with implantable defibrillators in patients
resuscitated from cardiac arrest : the Cardiac Arrest Study Hamburg (CASH). Circulation. 2000; 102:748-54.
134. Connolly SJ, Hallstrom AP, Cappato R, et al. Meta-analysis of the implantable cardioverter defibrillator secondary prevention trials. AVID,
CASH and CIDS studies. Antiarrhythmics vs Implantable Defibrillator study. Cardiac Arrest Study Hamburg . Canadian Implantable
Defibrillator Study. Eur. Heart J. 2000; 21:2071-8.
135. Lau EW, Griffith MJ, Pathmanathan RK, et al. The Midlands Trial of Empirical Amiodarone versus Electrophysiology-guided Interventions
and Implantable Cardioverter-defibrillators (MAVERIC): a multi-centre prospective randomised clinical trial on the secondary prevention of
sudden cardiac death. Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac
pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2004; 6:257-66.
136. Claro JC, Candia R, Rada G, et al. Amiodarone versus other pharmacological interventions for prevention of sudden cardiac death.
Cochrane Database Syst. Rev. 2015; 12:CD008093.
137. Raitt MH, Renfroe EG, Epstein AE, et al. "Stable" ventricular tachycardia is not a benign rhythm : insights from the antiarrhythmics versus
implantable defibrillators (AVID) registry. Circulation. 2001; 103:244-52.
138. Bass EB, Elson JJ, Fogoros RN, et al. Long-term prognosis of patients undergoing electrophysiologic studies for syncope of unknown origin.
Am J Cardiol. 1988; 62:1186-91.
139. Owens DK, Sanders GD, Heidenreich PA, et al. Effect of risk stratification on cost-effectiveness of the implantable cardioverter defibrillator.
Am Heart J. 2002; 144:440-8.
140. Ahn JM, Lee KH, Yoo SY, et al. Prognosis of variant angina manifesting as aborted sudden cardiac death. J Am Coll Cardiol. 2016; 68:137-45.
141. Yamashina Y, Yagi T, Namekawa A, et al. Favorable outcomes of patients with vasospastic angina associated with cardiac arrest. J Cardiol.
2014; 63:41-5.
142. Eschalier R, Souteyrand G, Jean F, et al. Should an implanted defibrillator be considered in patients with vasospastic angina? Arch.
Cardiovasc Dis. 2014; 107:42-7.
143. Matsue Y, Suzuki M, Nishizaki M, et al. Clinical implications of an implantable cardioverter-defibrillator in patients with vasospastic angina
and lethal ventricular arrhythmia. J Am Coll Cardiol. 2012; 60:908-13.
144. Takagi Y, Yasuda S, Tsunoda R, et al. Clinical characteristics and long-term prognosis of vasospastic angina patients who survived out-of-
hospital cardiac arrest: multicenter registry study of the Japanese Coronary Spasm Association. Circ. Arrhythm. Electrophysiol. 2011;
4:295-302.
145. Meisel SR, Mazur A, Chetboun I, et al. Usefulness of implantable cardioverter-defibrillators in refractory variant angina pectoris
complicated by ventricular fibrillation in patients with angiographically normal coronary arteries. Am. J. Cardiol. 2002; 89:1114-6.
146. Chevalier P, Dacosta A, Defaye P, et al. Arrhythmic cardiac arrest due to isolated coronary artery spasm: long-term outcome of seven
resuscitated patients. J Am Coll Cardiol. 1998; 31:57-61.
147. Myerburg RJ, Kessler KM, Mallon SM, et al. Life-threatening ventricular arrhythmias in patients with silent myocardial ischemia due to
coronary-artery spasm. N. Engl. J. Med. 1992; 326:1451-5.

329

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
148. Saxon LA, Wiener I, Natterson PD, et al. Monomorphic versus polymorphic ventricular tachycardia after coronary artery bypass grafting.
Am. J. Cardiol. 1995; 75:403-5.
149. Ascione R, Reeves BC, Santo K, et al. Predictors of new malignant ventricular arrhythmias after coronary surgery: a case-control study. J
Am Coll Cardiol. 2004; 43:1630-8.
150. Steinberg JS, Gaur A, Sciacca R, et al. New-onset sustained ventricular tachycardia after cardiac surgery. Circulation. 1999; 99:903-8.
151. Bigger JT, Jr. Prophylactic use of implanted cardiac defibrillators in patients at high risk for ventricular arrhythmias after coronary-artery
bypass graft surgery. Coronary Artery Bypass Graft (CABG) Patch Trial Investigators. N. Engl. J. Med. 1997; 337:1569-75.
152. Hohnloser SH, Kuck KH, Dorian P, et al. Prophylactic use of an implantable cardioverter-defibrillator after acute myocardial infarction. N.
Engl. J. Med. 2004; 351:2481-8.
153. Steinbeck G, Andresen D, Seidl K, et al. Defibrillator implantation early after myocardial infarction. N. Engl. J. Med. 2009; 361:1427-36.
154. Piccini JP, Berger JS and O'Connor CM. Amiodarone for the prevention of sudden cardiac death: a meta-analysis of randomized controlled
trials. Eur. Heart J. 2009; 30:1245-53.
155. Cantero-Perez EM, Sobrino-Marquez JM, Grande-Trillo A, et al. Implantable cardioverter defibrillator for primary prevention in patients
with severe ventricular dysfunction awaiting heart transplantation. Transplantation proceedings. 2013; 45:3659-61.
156. Frohlich GM, Holzmeister J, Hubler M, et al. Prophylactic implantable cardioverter defibrillator treatment in patients with end-stage heart
failure awaiting heart transplantation. Heart (British Cardiac Society). 2013; 99:1158-65.
157. Gandjbakhch E, Rovani M, Varnous S, et al. Implantable cardioverter-defibrillators in end-stage heart failure patients listed for heart
transplantation: Results from a large retrospective registry. Archives of cardiovascular diseases. 2016; 109:476-85.
158. Vakil K, Duval S, Cogswell R, et al. Impact of implantable cardioverter-defibrillators on waitlist mortality among patients awaiting heart
transplantation: an UNOS/OPTN analysis. JACCCEP. 2016.
159. Connolly SJ, Dorian P, Roberts RS, et al. Comparison of beta-blockers, amiodarone plus beta-blockers, or sotalol for prevention of shocks
from implantable cardioverter defibrillators: the OPTIC Study: a randomized trial. Jama. 2006; 295:165-71.
160. Pacifico A, Hohnloser SH, Williams JH, et al. Prevention of implantable-defibrillator shocks by treatment with sotalol. d,l-Sotalol
Implantable Cardioverter-Defibrillator Study Group. N. Engl. J. Med. 1999; 340:1855-62.
161. Kettering K, Mewis C, Dornberger V, et al. Efficacy of metoprolol and sotalol in the prevention of recurrences of sustained ventricular
tachyarrhythmias in patients with an implantable cardioverter defibrillator. Pacing Clin. Electrophysiol. 2002; 25:1571-6.
162. Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The Cardiac
Arrhythmia Suppression Trial. N. Engl. J. Med. 1991; 324:781-8.
163. Seidl K, Hauer B, Schwick NG, et al. Comparison of metoprolol and sotalol in preventing ventricular tachyarrhythmias after the
implantation of a cardioverter/defibrillator. Am. J. Cardiol. 1998; 82:744-8.
164. Kuhlkamp V, Mewis C, Mermi J, et al. Suppression of sustained ventricular tachyarrhythmias: a comparison of d,l-sotalol with no
antiarrhythmic drug treatment. J Am Coll Cardiol. 1999; 33:46-52.
165. Brodine WN, Tung RT, Lee JK, et al. Effects of beta-blockers on implantable cardioverter defibrillator therapy and survival in the patients
with ischemic cardiomyopathy (from the Multicenter Automatic Defibrillator Implantation Trial-II). Am. J. Cardiol. 2005; 96:691-5.
330

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
166. Reddy VY, Reynolds MR, Neuzil P, et al. Prophylactic catheter ablation for the prevention of defibrillator therapy. N. Engl. J. Med. 2007;
357:2657-65.
167. Sapp JL, Wells GA, Parkash R, et al. Ventricular tachycardia ablation versus escalation of antiarrhythmic drugs. N. Engl. J. Med. 2016;
375:111-21.
168. Kuck KH, Schaumann A, Eckardt L, et al. Catheter ablation of stable ventricular tachycardia before defibrillator implantation in patients
with coronary heart disease (VTACH): a multicentre randomised controlled trial. Lancet (London, England). 2010; 375:31-40.
169. Al-Khatib SM, Daubert JP, Anstrom KJ, et al. Catheter ablation for ventricular tachycardia in patients with an implantable cardioverter
defibrillator (CALYPSO) pilot trial. Journal of cardiovascular electrophysiology. 2015; 26:151-7.
170. Blanck Z, Dhala A, Deshpande S, et al. Bundle branch reentrant ventricular tachycardia: cumulative experience in 48 patients. J. Cardiovasc.
Electrophysiol. 1993; 4:253-62.
171. Brugada J, Aguinaga L, Mont L, et al. Coronary artery revascularization in patients with sustained ventricular arrhythmias in the chronic
phase of a myocardial infarction: effects on the electrophysiologic substrate and outcome. J Am Coll Cardiol. 2001; 37:529-33.
172. Sears SF, Jr., Todaro JF, Lewis TS, et al. Examining the psychosocial impact of implantable cardioverter defibrillators: a literature review.
Clinical cardiology. 1999; 22:481-9.
173. Lopera G, Stevenson WG, Soejima K, et al. Identification and ablation of three types of ventricular tachycardia involving the his-purkinje
system in patients with heart disease. J. Cardiovasc. Electrophysiol. 2004; 15:52-8.
174. Mehdirad AA, Keim S, Rist K, et al. Long-term clinical outcome of right bundle branch radiofrequency catheter ablation for treatment of
bundle branch reentrant ventricular tachycardia. Pacing Clin. Electrophysiol. 1995; 18:2135-43.
175. Dinov B, Fiedler L, Schonbauer R, et al. Outcomes in catheter ablation of ventricular tachycardia in dilated nonischemic cardiomyopathy
compared with ischemic cardiomyopathy: results from the Prospective Heart Centre of Leipzig VT (HELP-VT) Study. Circulation. 2014;
129:728-36.
176. Tanner H, Hindricks G, Volkmer M, et al. Catheter ablation of recurrent scar-related ventricular tachycardia using electroanatomical
mapping and irrigated ablation technology: results of the prospective multicenter Euro-VT-study. J. Cardiovasc. Electrophysiol. 2010;
21:47-53.
177. Marchlinski FE, Haffajee CI, Beshai JF, et al. Long-term success of irrigated radiofrequency catheter ablation of sustained ventricular
tachycardia: post-approval THERMOCOOL VT trial. J Am Coll Cardiol. 2016; 67:674-83.
178. Tung R, Vaseghi M, Frankel DS, et al. Freedom from recurrent ventricular tachycardia after catheter ablation is associated with improved
survival in patients with structural heart disease: An International VT Ablation Center Collaborative Group study. Heart rhythm. 2015;
12:1997-2007.
179. Mallidi J, Nadkarni GN, Berger RD, et al. Meta-analysis of catheter ablation as an adjunct to medical therapy for treatment of ventricular
tachycardia in patients with structural heart disease. Heart rhythm. 2011; 8:503-10.
180. Calkins H, Epstein A, Packer D, et al. Catheter ablation of ventricular tachycardia in patients with structural heart disease using cooled
radiofrequency energy: results of a prospective multicenter study. Cooled RF Multi Center Investigators Group. J Am Coll Cardiol. 2000;
35:1905-14.
331

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
181. Stevenson WG, Wilber DJ, Natale A, et al. Irrigated radiofrequency catheter ablation guided by electroanatomic mapping for recurrent
ventricular tachycardia after myocardial infarction: the multicenter thermocool ventricular tachycardia ablation trial. Circulation. 2008;
118:2773-82.
182. Ackerman MJ, Priori SG, Willems S, et al. HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies
and cardiomyopathies: this document was developed as a partnership between the Heart Rhythm Society (HRS) and the European Heart
Rhythm Association (EHRA). Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on
cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2011; 13:1077-109.
183. American Geriatrics Society Expert Panel on the Care of Older Adults with M. Guiding Principles for the Care of Older Adults with
Multimorbidity: An Approach for Clinicians. Journal of the American Geriatrics Society. 2012; 60:E1-E25.
184. Hershberger RE, Morales A and Siegfried JD. Clinical and genetic issues in dilated cardiomyopathy: a review for genetics professionals.
Genet. Med. 2010; 12:655-67.
185. Piers SR, Tao Q, CF vHvT, et al. Contrast-enhanced MRI-derived scar patterns and associated ventricular tachycardias in nonischemic
cardiomyopathy: implications for the ablation strategy. Circ. Arrhythm. Electrophysiol. 2013; 6:875-83.
186. Greulich S, Deluigi CC, Gloekler S, et al. CMR imaging predicts death and other adverse events in suspected cardiac sarcoidosis. JACC.
Cardiovasc. Imaging. 2013; 6:501-11.
187. Kuruvilla S, Adenaw N, Katwal AB, et al. Late gadolinium enhancement on cardiac magnetic resonance predicts adverse cardiovascular
outcomes in nonischemic cardiomyopathy: a systematic review and meta-analysis. Circ. Cardiovasc. Imaging. 2014; 7:250-8.
188. Tokuda M, Tedrow UB, Kojodjojo P, et al. Catheter ablation of ventricular tachycardia in nonischemic heart disease. Circ Arrhythm
Electrophysiol. 2012; 5:992-1000.
189. Oloriz T, Silberbauer J, Maccabelli G, et al. Catheter ablation of ventricular arrhythmia in nonischemic cardiomyopathy: anteroseptal versus
inferolateral scar sub-types. Circ Arrhythm Electrophysiol. 2014; 7:414-23.
190. Proietti R, Essebag V, Beardsall J, et al. Substrate-guided ablation of haemodynamically tolerated and untolerated ventricular tachycardia
in patients with structural heart disease: effect of cardiomyopathy type and acute success on long-term outcome. Europace : European
pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular
electrophysiology of the European Society of Cardiology. 2015; 17:461-7.
191. Haqqani HM, Tschabrunn CM, Tzou WS, et al. Isolated septal substrate for ventricular tachycardia in nonischemic dilated cardiomyopathy:
incidence, characterization, and implications. Heart rhythm. 2011; 8:1169-76.
192. Kuhne M, Abrams G, Sarrazin JF, et al. Isolated potentials and pace-mapping as guides for ablation of ventricular tachycardia in various
types of nonischemic cardiomyopathy. Journal of cardiovascular electrophysiology. 2010; 21:1017-23.
193. Cano O, Hutchinson M, Lin D, et al. Electroanatomic substrate and ablation outcome for suspected epicardial ventricular tachycardia in left
ventricular nonischemic cardiomyopathy. J Am Coll Cardiol. 2009; 54:799-808.
194. Delacretaz E, Stevenson WG, Ellison KE, et al. Mapping and radiofrequency catheter ablation of the three types of sustained monomorphic
ventricular tachycardia in nonischemic heart disease. Journal of cardiovascular electrophysiology. 2000; 11:11-7.

332

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
195. Desai AS, Fang JC, Maisel WH, et al. Implantable defibrillators for the prevention of mortality in patients with nonischemic
cardiomyopathy: a meta-analysis of randomized controlled trials. Jama. 2004; 292:2874-9.
196. Ruwald MH, Okumura K, Kimura T, et al. Syncope in high-risk cardiomyopathy patients with implantable defibrillators: frequency, risk
factors, mechanisms, and association with mortality: results from the multicenter automatic defibrillator implantation trial-reduce
inappropriate therapy (MADIT-RIT) study. Circulation. 2014; 129:545-52.
197. Knight BP, Goyal R, Pelosi F, et al. Outcome of patients with nonischemic dilated cardiomyopathy and unexplained syncope treated with an
implantable defibrillator. J Am Coll Cardiol. 1999; 33:1964-70.
198. Brilakis ES, Shen WK, Hammill SC, et al. Role of programmed ventricular stimulation and implantable cardioverter defibrillators in patients
with idiopathic dilated cardiomyopathy and syncope. Pacing Clin. Electrophysiol. 2001; 24:1623-30.
199. Fonarow GC, Feliciano Z, Boyle NG, et al. Improved survival in patients with nonischemic advanced heart failure and syncope treated with
an implantable cardioverter-defibrillator. Am. J. Cardiol. 2000; 85:981-5.
200. Olshansky B, Poole JE, Johnson G, et al. Syncope predicts the outcome of cardiomyopathy patients: analysis of the SCD-HeFT study. J Am
Coll Cardiol. 2008; 51:1277-82.
201. Bnsch D, Antz M, Boczor S, et al. Primary prevention of sudden cardiac death in idiopathic dilated cardiomyopathy: the Cardiomyopathy
Trial (CAT). Circulation. 2002; 105:1453-8.
202. Strickberger SA, Hummel JD, Bartlett TG, et al. Amiodarone versus implantable cardioverter-defibrillator:randomized trial in patients with
nonischemic dilated cardiomyopathy and asymptomatic nonsustained ventricular tachycardia--AMIOVIRT. J Am Coll Cardiol. 2003;
41:1707-12.
203. Kadish A, Dyer A, Daubert JP, et al. Prophylactic defibrillator implantation in patients with nonischemic dilated cardiomyopathy. N. Engl. J.
Med. 2004; 350:2151-8.
204. Bristow MR, Saxon LA, Boehmer J, et al. Cardiac-resynchronization therapy with or without an implantable defibrillator in advanced
chronic heart failure. N. Engl. J. Med. 2004; 350:2140-50.
205. Kober L, Bloch Thomsen PE, Moller M, et al. Effect of dofetilide in patients with recent myocardial infarction and left-ventricular
dysfunction: a randomised trial. Lancet (London, England). 2000; 356:2052-8.
206. Grimm W, Christ M, Bach J, et al. Noninvasive arrhythmia risk stratification in idiopathic dilated cardiomyopathy: results of the Marburg
Cardiomyopathy Study. Circulation. 2003; 108:2883-91.
207. Goldberger JJ, Subacius H, Patel T, et al. Sudden cardiac death risk stratification in patients with nonischemic dilated cardiomyopathy. J Am
Coll Cardiol. 2014; 63:1879-89.
208. Anselme F, Moubarak G, Savoure A, et al. Implantable cardioverter-defibrillators in lamin A/C mutation carriers with cardiac conduction
disorders. Heart rhythm. 2013; 10:1492-8.
209. van Rijsingen IA, Arbustini E, Elliott PM, et al. Risk factors for malignant ventricular arrhythmias in lamin a/c mutation carriers a European
cohort study. J Am Coll Cardiol. 2012; 59:493-500.
210. Pasotti M, Klersy C, Pilotto A, et al. Long-term outcome and risk stratification in dilated cardiolaminopathies. J Am Coll Cardiol. 2008;
52:1250-60.
333

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
211. Van Berlo JH, de Voogt WG, van der Kooi AJ, et al. Meta-analysis of clinical characteristics of 299 carriers of LMNA gene mutations: do
lamin A/C mutations portend a high risk of sudden death? J. Mol. Med. (Berl. ). 2005; 83:79-83.
212. Kutyifa V, Moss AJ, Klein H, et al. Use of the wearable cardioverter defibrillator in high-risk cardiac patients: data from the Prospective
Registry of Patients Using the Wearable Cardioverter Defibrillator (WEARIT-II Registry). Circulation. 2015; 132:1613-9.
213. Singh M, Wang NC, Jain S, et al. Utility of the wearable cardioverter-defibrillator in patients with newly diagnosed cardiomyopathy: a
decade-long single-center experience. J Am Coll Cardiol. 2015; 66:2607-13.
214. Uyei J and Braithwaite RS. Effectiveness of wearable defibrillators: systematic review and quality of evidence. Int. J Technol. Assess. Health
Care. 2014; 30:194-202.
215. Al-Khatib SM, Fonarow GC, Joglar JA, et al. Primary prevention implantable cardioverter defibillators in patients with nonischemic
cardiomyopathy: A meta-analysis. JAMA Cardiology. 2017.
216. Quarta G, Muir A, Pantazis A, et al. Familial evaluation in arrhythmogenic right ventricular cardiomyopathy: impact of genetics and revised
task force criteria. Circulation. 2011; 123:2701-9.
217. Kapplinger JD, Landstrom AP, Salisbury BA, et al. Distinguishing arrhythmogenic right ventricular cardiomyopathy/dysplasia-associated
mutations from background genetic noise. J Am Coll Cardiol. 2011; 57:2317-27.
218. Bhonsale A, James CA, Tichnell C, et al. Risk stratification in arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated
desmosomal mutation carriers. Circ. Arrhythm. Electrophysiol. 2013; 6:569-78.
219. Marcus FI, Edson S and Towbin JA. Genetics of arrhythmogenic right ventricular cardiomyopathy: a practical guide for physicians. J Am Coll
Cardiol. 2013; 61:1945-8.
220. Bhonsale A, Groeneweg JA, James CA, et al. Impact of genotype on clinical course in arrhythmogenic right ventricular
dysplasia/cardiomyopathy-associated mutation carriers. Eur. Heart J. 2015; 36:847-55.
221. Rigato I, Bauce B, Rampazzo A, et al. Compound and digenic heterozygosity predicts lifetime arrhythmic outcome and sudden cardiac
death in desmosomal gene-related arrhythmogenic right ventricular cardiomyopathy. Circ. Cardiovasc. Genet. 2013; 6:533-42.
222. Groeneweg JA, Bhonsale A, James CA, et al. Clinical Presentation, Long-Term Follow-Up, and Outcomes of 1001 Arrhythmogenic Right
Ventricular Dysplasia/Cardiomyopathy Patients and Family Members. Circ. Cardiovasc. Genet. 2015; 8:437-46.
223. te Riele AS, James CA, Groeneweg JA, et al. Approach to family screening in arrhythmogenic right ventricular dysplasia/cardiomyopathy.
Eur. Heart J. 2016; 37:755-63.
224. Kamath GS, Zareba W, Delaney J, et al. Value of the signal-averaged electrocardiogram in arrhythmogenic right ventricular
cardiomyopathy/dysplasia. Heart rhythm. 2011; 8:256-62.
225. Marcus FI, Fontaine GH, Guiraudon G, et al. Right ventricular dysplasia: a report of 24 adult cases. Circulation. 1982; 65:384-98.
226. Corrado D, Basso C, Thiene G, et al. Spectrum of clinicopathologic manifestations of arrhythmogenic right ventricular
cardiomyopathy/dysplasia: a multicenter study. J Am Coll Cardiol. 1997; 30:1512-20.
227. Link MS, Laidlaw D, Polonsky B, et al. Ventricular arrhythmias in the North American multidisciplinary study of ARVC: predictors,
characteristics, and treatment. J Am Coll Cardiol. 2014; 64:119-25.

334

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
228. Corrado D, Wichter T, Link MS, et al. Treatment of arrhythmogenic right ventricular cardiomyopathy/dysplasia: an international task force
consensus statement. Circulation. 2015; 132:441-53.
229. Corrado D, Leoni L, Link MS, et al. Implantable cardioverter-defibrillator therapy for prevention of sudden death in patients with
arrhythmogenic right ventricular cardiomyopathy/dysplasia. Circulation. 2003; 108:3084-91.
230. Piccini JP, Dalal D, Roguin A, et al. Predictors of appropriate implantable defibrillator therapies in patients with arrhythmogenic right
ventricular dysplasia. Heart rhythm. 2005; 2:1188-94.
231. Bhonsale A, James CA, Tichnell C, et al. Incidence and predictors of implantable cardioverter-defibrillator therapy in patients with
arrhythmogenic right ventricular dysplasia/cardiomyopathy undergoing implantable cardioverter-defibrillator implantation for primary
prevention. J Am Coll Cardiol. 2011; 58:1485-96.
232. Dalal D, Jain R, Tandri H, et al. Long-term efficacy of catheter ablation of ventricular tachycardia in patients with arrhythmogenic right
ventricular dysplasia/cardiomyopathy. J Am Coll Cardiol. 2007; 50:432-40.
233. Garcia FC, Bazan V, Zado ES, et al. Epicardial substrate and outcome with epicardial ablation of ventricular tachycardia in arrhythmogenic
right ventricular cardiomyopathy/dysplasia. Circulation. 2009; 120:366-75.
234. Philips B, Madhavan S, James C, et al. Outcomes of catheter ablation of ventricular tachycardia in arrhythmogenic right ventricular
dysplasia/cardiomyopathy. Circ. Arrhythm. Electrophysiol. 2012; 5:499-505.
235. Bai R, Di BL, Shivkumar K, et al. Ablation of ventricular arrhythmias in arrhythmogenic right ventricular dysplasia/cardiomyopathy:
arrhythmia-free survival after endo-epicardial substrate based mapping and ablation. Circ. Arrhythm. Electrophysiol. 2011; 4:478-85.
236. Berruezo A, Fernandez-Armenta J, Mont L, et al. Combined endocardial and epicardial catheter ablation in arrhythmogenic right
ventricular dysplasia incorporating scar dechanneling technique. Circ. Arrhythm. Electrophysiol. 2012; 5:111-21.
237. Philips B, te Riele AS, Sawant A, et al. Outcomes and ventricular tachycardia recurrence characteristics after epicardial ablation of
ventricular tachycardia in arrhythmogenic right ventricular dysplasia/cardiomyopathy. Heart rhythm. 2015; 12:716-25.
238. Santangeli P, Zado ES, Supple GE, et al. Long-term outcome with catheter ablation of ventricular tachycardia in patients with
arrhythmogenic right ventricular cardiomyopathy. Circ. Arrhythm. Electrophysiol. 2015; 8:1413-21.
239. James CA, Bhonsale A, Tichnell C, et al. Exercise increases age-related penetrance and arrhythmic risk in arrhythmogenic right ventricular
dysplasia/cardiomyopathy-associated desmosomal mutation carriers. J Am Coll Cardiol. 2013; 62:1290-7.
240. Sawant AC, Bhonsale A, te Riele AS, et al. Exercise has a disproportionate role in the pathogenesis of arrhythmogenic right ventricular
dysplasia/cardiomyopathy in patients without desmosomal mutations. Journal of the American Heart Association. 2014; 3:e001471.
241. Ruwald AC, Marcus F, Estes NA, 3rd, et al. Association of competitive and recreational sport participation with cardiac events in patients
with arrhythmogenic right ventricular cardiomyopathy: results from the North American multidisciplinary study of arrhythmogenic right
ventricular cardiomyopathy. European heart journal. 2015; 36:1735-43.
242. Sawant AC, te Riele AS, Tichnell C, et al. Safety of American Heart Association-recommended minimum exercise for desmosomal mutation
carriers. Heart rhythm. 2016; 13:199-207.
243. Saberniak J, Hasselberg NE, Borgquist R, et al. Vigorous physical activity impairs myocardial function in patients with arrhythmogenic right
ventricular cardiomyopathy and in mutation positive family members. Eur. J Heart Fail. 2014; 16:1337-44.
335

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
244. Sen-Chowdhry S, Syrris P, Prasad SK, et al. Left-dominant arrhythmogenic cardiomyopathy: an under-recognized clinical entity. J Am Coll
Cardiol. 2008; 52:2175-87.
245. Vermes E, Strohm O, Otmani A, et al. Impact of the revision of arrhythmogenic right ventricular cardiomyopathy/dysplasia task force
criteria on its prevalence by CMR criteria. JACC. Cardiovascular imaging. 2011; 4:282-7.
246. te Riele AS, Bhonsale A, James CA, et al. Incremental value of cardiac magnetic resonance imaging in arrhythmic risk stratification of
arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated desmosomal mutation carriers. J Am Coll Cardiol. 2013; 62:1761-9.
247. Te Riele AS, James CA, Philips B, et al. Mutation-positive arrhythmogenic right ventricular dysplasia/cardiomyopathy: the triangle of
dysplasia displaced. Journal of cardiovascular electrophysiology. 2013; 24:1311-20.
248. Liu T, Pursnani A, Sharma UC, et al. Effect of the 2010 task force criteria on reclassification of cardiovascular magnetic resonance criteria
for arrhythmogenic right ventricular cardiomyopathy. Journal of cardiovascular magnetic resonance : official journal of the Society for
Cardiovascular Magnetic Resonance. 2014; 16:47.
249. Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification
of the task force criteria. Circulation. 2010; 121:1533-41.
250. Hershberger RE, Lindenfeld J, Mestroni L, et al. Genetic evaluation of cardiomyopathy--a Heart Failure Society of America practice
guideline. Journal of cardiac failure. 2009; 15:83-97.
251. Maron BJ, Shen WK, Link MS, et al. Efficacy of implantable cardioverter-defibrillators for the prevention of sudden death in patients with
hypertrophic cardiomyopathy. N. Engl. J. Med. 2000; 342:365-73.
252. Christiaans I, van Langen IM, Birnie E, et al. Genetic counseling and cardiac care in predictively tested hypertrophic cardiomyopathy
mutation carriers: the patients' perspective. American journal of medical genetics. Part A. 2009; 149a:1444-51.
253. Hamang A, Eide GE, Rokne B, et al. Predictors of heart-focused anxiety in patients undergoing genetic investigation and counseling of long
QT syndrome or hypertrophic cardiomyopathy: a one year follow-up. Journal of genetic counseling. 2012; 21:72-84.
254. Bos JM, Will ML, Gersh BJ, et al. Characterization of a phenotype-based genetic test prediction score for unrelated patients with
hypertrophic cardiomyopathy. Mayo Clinic proceedings. 2014; 89:727-37.
255. O'Mahony C, Jichi F, Pavlou M, et al. A novel clinical risk prediction model for sudden cardiac death in hypertrophic cardiomyopathy (HCM
risk-SCD). Eur. Heart J. 2014; 35:2010-20.
256. Elliott PM, Sharma S, Varnava A, et al. Survival after cardiac arrest or sustained ventricular tachycardia in patients with hypertrophic
cardiomyopathy. J Am Coll Cardiol. 1999; 33:1596-601.
257. Maron BJ, Spirito P, Shen WK, et al. Implantable cardioverter-defibrillators and prevention of sudden cardiac death in hypertrophic
cardiomyopathy. Jama. 2007; 298:405-12.
258. Lin G, Nishimura RA, Gersh BJ, et al. Device complications and inappropriate implantable cardioverter defibrillator shocks in patients with
hypertrophic cardiomyopathy. Heart (British Cardiac Society). 2009; 95:709-14.
259. Syska P, Przybylski A, Chojnowska L, et al. Implantable cardioverter-defibrillator in patients with hypertrophic cardiomyopathy: efficacy
and complications of the therapy in long-term follow-up. J. Cardiovasc. Electrophysiol. 2010; 21:883-9.

336

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
260. O'Mahony C, Lambiase PD, Quarta G, et al. The long-term survival and the risks and benefits of implantable cardioverter defibrillators in
patients with hypertrophic cardiomyopathy. Heart (British Cardiac Society). 2012; 98:116-25.
261. Melacini P, Maron BJ, Bobbo F, et al. Evidence that pharmacological strategies lack efficacy for the prevention of sudden death in
hypertrophic cardiomyopathy. Heart (British Cardiac Society). 2007; 93:708-10.
262. McKenna WJ, Oakley CM, Krikler DM, et al. Improved survival with amiodarone in patients with hypertrophic cardiomyopathy and
ventricular tachycardia. Br. Heart J. 1985; 53:412-6.
263. Olivotto I, Maron BJ, Montereggi A, et al. Prognostic value of systemic blood pressure response during exercise in a community-based
patient population with hypertrophic cardiomyopathy. J Am Coll Cardiol. 1999; 33:2044-51.
264. Sadoul N, Prasad K, Elliott PM, et al. Prospective prognostic assessment of blood pressure response during exercise in patients with
hypertrophic cardiomyopathy. Circulation. 1997; 96:2987-91.
265. Sorajja P, Nishimura RA, Ommen SR, et al. Use of echocardiography in patients with hypertrophic cardiomyopathy: clinical implications of
massive hypertrophy. J Am Soc. Echocardiogr. 2006; 19:788-95.
266. Maki S, Ikeda H, Muro A, et al. Predictors of sudden cardiac death in hypertrophic cardiomyopathy. Am. J. Cardiol. 1998; 82:774-8.
267. Elliott PM, Gimeno JR, Tome MT, et al. Left ventricular outflow tract obstruction and sudden death risk in patients with hypertrophic
cardiomyopathy. Eur. Heart J. 2006; 27:1933-41.
268. Monserrat L, Elliott PM, Gimeno JR, et al. Non-sustained ventricular tachycardia in hypertrophic cardiomyopathy: an independent marker
of sudden death risk in young patients. J Am Coll Cardiol. 2003; 42:873-9.
269. Spirito P, Bellone P, Harris KM, et al. Magnitude of left ventricular hypertrophy and risk of sudden death in hypertrophic cardiomyopathy.
N. Engl. J. Med. 2000; 342:1778-85.
270. Elliott PM, Gimeno Blanes JR, Mahon NG, et al. Relation between severity of left-ventricular hypertrophy and prognosis in patients with
hypertrophic cardiomyopathy. Lancet (London, England). 2001; 357:420-4.
271. Elliott PM, Poloniecki J, Dickie S, et al. Sudden death in hypertrophic cardiomyopathy: identification of high risk patients. J Am Coll Cardiol.
2000; 36:2212-8.
272. Ackerman MJ, VanDriest SL, Ommen SR, et al. Prevalence and age-dependence of malignant mutations in the beta-myosin heavy chain and
troponin T genes in hypertrophic cardiomyopathy: a comprehensive outpatient perspective. J Am Coll Cardiol. 2002; 39:2042-8.
273. Lopes LR, Rahman MS and Elliott PM. A systematic review and meta-analysis of genotype-phenotype associations in patients with
hypertrophic cardiomyopathy caused by sarcomeric protein mutations. Heart (British Cardiac Society). 2013; 99:1800-11.
274. Bos JM, Maron BJ, Ackerman MJ, et al. Role of family history of sudden death in risk stratification and prevention of sudden death with
implantable defibrillators in hypertrophic cardiomyopathy. Am. J. Cardiol. 2010; 106:1481-6.
275. Spirito P, Autore C, Rapezzi C, et al. Syncope and risk of sudden death in hypertrophic cardiomyopathy. Circulation. 2009; 119:1703-10.
276. Maron BJ, Doerer JJ, Haas TS, et al. Sudden deaths in young competitive athletes: analysis of 1866 deaths in the United States, 1980-2006.
Circulation. 2009; 119:1085-92.
277. Kuck KH, Kunze KP, Schluter M, et al. Programmed electrical stimulation in hypertrophic cardiomyopathy. Results in patients with and
without cardiac arrest or syncope. Eur. Heart J. 1988; 9:177-85.
337

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
278. Zhu DW, Sun H, Hill R, et al. The value of electrophysiology study and prophylactic implantation of cardioverter defibrillator in patients
with hypertrophic cardiomyopathy. Pacing Clin. Electrophysiol. 1998; 21:299-302.
279. Christiaans I, Birnie E, van Langen IM, et al. The yield of risk stratification for sudden cardiac death in hypertrophic cardiomyopathy
myosin-binding protein C gene mutation carriers: focus on predictive screening. Eur. Heart J. 2010; 31:842-8.
280. Olivotto I, Girolami F, Ackerman MJ, et al. Myofilament protein gene mutation screening and outcome of patients with hypertrophic
cardiomyopathy. Mayo Clin. Proc. 2008; 83:630-8.
281. Ingles J, Sarina T, Yeates L, et al. Clinical predictors of genetic testing outcomes in hypertrophic cardiomyopathy. Genet. Med. 2013;
15:972-7.
282. Jensen MK, Havndrup O, Christiansen M, et al. Penetrance of hypertrophic cardiomyopathy in children and adolescents: a 12-year follow-
up study of clinical screening and predictive genetic testing. Circulation. 2013; 127:48-54.
283. Girolami F, Ho CY, Semsarian C, et al. Clinical features and outcome of hypertrophic cardiomyopathy associated with triple sarcomere
protein gene mutations. J Am Coll Cardiol. 2010; 55:1444-53.
284. Klues HG, Schiffers A and Maron BJ. Phenotypic spectrum and patterns of left ventricular hypertrophy in hypertrophic cardiomyopathy:
morphologic observations and significance as assessed by two-dimensional echocardiography in 600 patients. J Am Coll Cardiol. 1995;
26:1699-708.
285. Adabag AS, Kuskowski MA and Maron BJ. Determinants for clinical diagnosis of hypertrophic cardiomyopathy. Am J Cardiol. 2006; 98:1507-
11.
286. Cooper LT, Jr., Berry GJ and Shabetai R. Idiopathic giant-cell myocarditis--natural history and treatment. Multicenter Giant Cell Myocarditis
Study Group Investigators. N. Engl. J. Med. 1997; 336:1860-6.
287. Kandolin R, Lehtonen J, Salmenkivi K, et al. Diagnosis, treatment, and outcome of giant-cell myocarditis in the era of combined
immunosuppression. Circ. Heart Fail. 2013; 6:15-22.
288. Maleszewski JJ, Orellana VM, Hodge DO, et al. Long-term risk of recurrence, morbidity and mortality in giant cell myocarditis. Am. J.
Cardiol. 2015; 115:1733-8.
289. Feldman AM, Klein H, Tchou P, et al. Use of a wearable defibrillator in terminating tachyarrhythmias in patients at high risk for sudden
death: results of the WEARIT/BIROAD. Pacing Clin. Electrophysiol. 2004; 27:4-9.
290. Kao AC, Krause SW, Handa R, et al. Wearable defibrillator use in heart failure (WIF): results of a prospective registry. BMC Cardiovasc
Disord. 2012; 12:123.
291. Naruse Y, Sekiguchi Y, Nogami A, et al. Systematic treatment approach to ventricular tachycardia in cardiac sarcoidosis. Circ. Arrhythm.
Electrophysiol. 2014; 7:407-13.
292. Takaya Y, Kusano KF, Nakamura K, et al. Outcomes in patients with high-degree atrioventricular block as the initial manifestation of cardiac
sarcoidosis. Am. J. Cardiol. 2015; 115:505-9.
293. Kandolin R, Lehtonen J, Airaksinen J, et al. Cardiac sarcoidosis: epidemiology, characteristics, and outcome over 25 years in a nationwide
study. Circulation. 2015; 131:624-32.

338

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
294. Yazaki Y, Isobe M, Hiroe M, et al. Prognostic determinants of long-term survival in Japanese patients with cardiac sarcoidosis treated with
prednisone. Am. J. Cardiol. 2001; 88:1006-10.
295. Aizer A, Stern EH, Gomes JA, et al. Usefulness of programmed ventricular stimulation in predicting future arrhythmic events in patients
with cardiac sarcoidosis. Am. J. Cardiol. 2005; 96:276-82.
296. Mehta D, Mori N, Goldbarg SH, et al. Primary prevention of sudden cardiac death in silent cardiac sarcoidosis: role of programmed
ventricular stimulation. Circ. Arrhythm. Electrophysiol. 2011; 4:43-8.
297. Coleman GC, Shaw PW, Balfour PC, Jr., et al. Prognostic value of myocardial scarring on CMR in patients with cardiac sarcoidosis: a
systematic review and meta-analysis. JACC. Cardiovasc. Imaging. 2016.
298. Heng EL, Bolger AP, Kempny A, et al. Neurohormonal activation and its relation to outcomes late after repair of tetralogy of Fallot. Heart
(British Cardiac Society). 2015; 101:447-54.
299. Murtagh G, Laffin LJ, Beshai JF, et al. Prognosis of myocardial damage in sarcoidosis patients with preserved left ventricular ejection
fraction: risk stratification using cardiovascular magnetic resonance. Circ. Cardiovasc. Imaging. 2016; 9:e003738.
300. Crawford T, Mueller G, Sarsam S, et al. Magnetic resonance imaging for identifying patients with cardiac sarcoidosis and preserved or
mildly reduced left ventricular function at risk of ventricular arrhythmias. Circ. Arrhythm. Electrophysiol. 2014; 7:1109-15.
301. Blankstein R, Osborne M, Naya M, et al. Cardiac positron emission tomography enhances prognostic assessments of patients with
suspected cardiac sarcoidosis. J Am Coll Cardiol. 2014; 63:329-36.
302. Kron J, Sauer W, Schuller J, et al. Efficacy and safety of implantable cardiac defibrillators for treatment of ventricular arrhythmias in
patients with cardiac sarcoidosis. Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups
on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2013; 15:347-54.
303. Mohsen A, Jimenez A, Hood RE, et al. Cardiac sarcoidosis: electrophysiological outcomes on long-term follow-up and the role of the
implantable cardioverter-defibrillator. J. Cardiovasc. Electrophysiol. 2014; 25:171-6.
304. Schuller JL, Zipse M, Crawford T, et al. Implantable cardioverter defibrillator therapy in patients with cardiac sarcoidosis. J. Cardiovasc.
Electrophysiol. 2012; 23:925-9.
305. Yodogawa K, Seino Y, Ohara T, et al. Effect of corticosteroid therapy on ventricular arrhythmias in patients with cardiac sarcoidosis. Ann.
Noninvasive. Electrocardiol. 2011; 16:140-7.
306. Segawa M, Fukuda K, Nakano M, et al. Time course and factors correlating with ventricular tachyarrhythmias after introduction of steroid
therapy in cardiac sarcoidosis. Circ. Arrhythm. Electrophysiol. 2016; 9:e003353.
307. Varr BC, Zarafshar S, Coakley T, et al. Implantable cardioverter-defibrillator placement in patients with cardiac amyloidosis. Heart rhythm.
2014; 11:158-62.
308. Kristen AV, Dengler TJ, Hegenbart U, et al. Prophylactic implantation of cardioverter-defibrillator in patients with severe cardiac
amyloidosis and high risk for sudden cardiac death. Heart rhythm. 2008; 5:235-40.
309. Lubitz SA, Goldbarg SH and Mehta D. Sudden cardiac death in infiltrative cardiomyopathies: sarcoidosis, scleroderma, amyloidosis,
hemachromatosis. Prog. Cardiovasc Dis. 2008; 51:58-73.

339

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
310. Sandner SE, Wieselthaler G, Zuckermann A, et al. Survival benefit of the implantable cardioverter-defibrillator in patients on the waiting
list for cardiac transplantation. Circulation. 2001; 104:I171-6.
311. Opreanu M, Wan C, Singh V, et al. Wearable cardioverter-defibrillator as a bridge to cardiac transplantation: A national database analysis.
The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2015;
34:1305-9.
312. Vakil K, Kazmirczak F, Sathnur N, et al. Implantable cardioverter-defibrillator use in patients with left ventricular assist devices: a
systematic review and meta-analysis. JACC. Heart Fail. 2016; 4:772-9.
313. Tsai VW, Cooper J, Garan H, et al. The efficacy of implantable cardioverter-defibrillators in heart transplant recipients: results from a
multicenter registry. Circ. Heart Fail. 2009; 2:197-201.
314. McDowell DL and Hauptman PJ. Implantable defibrillators and cardiac resynchronization therapy in heart transplant recipients: results of a
national survey. The Journal of heart and lung transplantation : the official publication of the International Society for Heart
Transplantation. 2009; 28:847-50.
315. Neylon A, Canniffe C, Parlon B, et al. Implantable cardioverter-defibrillators in a heart transplant population: A single-center experience.
The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2016; 35:682-
4.
316. Tanawuttiwat T, Wagner KR, Tomaselli G, et al. Left ventricular dysfunction and conduction disturbances in patients with myotonic
muscular dystrophy type i and ii. JAMA Cardiology. 2017; 2:225-8.
317. Merino JL, Carmona JR, Fernandez-Lozano I, et al. Mechanisms of sustained ventricular tachycardia in myotonic dystrophy: implications for
catheter ablation. Circulation. 1998; 98:541-6.
318. Diegoli M, Grasso M, Favalli V, et al. Diagnostic work-up and risk stratification in X-linked dilated cardiomyopathies caused by dystrophin
defects. J Am Coll Cardiol. 2011; 58:925-34.
319. Meune C, Van Berlo JH, Anselme F, et al. Primary prevention of sudden death in patients with lamin A/C gene mutations. N. Engl. J. Med.
2006; 354:209-10.
320. Lallemand B, Clementy N, Bernard-Brunet A, et al. The evolution of infrahissian conduction time in myotonic dystrophy patients: clinical
implications. Heart (British Cardiac Society). 2012; 98:291-6.
321. Wahbi K, Meune C, Porcher R, et al. Electrophysiological study with prophylactic pacing and survival in adults with myotonic dystrophy and
conduction system disease. Jama. 2012; 307:1292-301.
322. McMurray JJV, stergren J, Swedberg K, et al. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular
systolic function taking angiotensin-converting-enzyme inhibitors: the CHARM-Added trial. The Lancet. 362:767-71.
323. Ha AH, Tarnopolsky MA, Bergstra TG, et al. Predictors of atrio-ventricular conduction disease, long-term outcomes in patients with
myotonic dystrophy types I and II. Pacing Clin. Electrophysiol. 2012; 35:1262-9.
324. Laurent V, Pellieux S, Corcia P, et al. Mortality in myotonic dystrophy patients in the area of prophylactic pacing devices. Int. J. Cardiol.
2011; 150:54-8.

340

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
325. Bhakta D, Shen C, Kron J, et al. Pacemaker and implantable cardioverter-defibrillator use in a US myotonic dystrophy type 1 population. J.
Cardiovasc. Electrophysiol. 2011; 22:1369-75.
326. Nazarian S, Wagner KR, Caffo BS, et al. Clinical predictors of conduction disease progression in type I myotonic muscular dystrophy. Pacing
Clin. Electrophysiol. 2011; 34:171-6.
327. Bhakta D, Groh MR, Shen C, et al. Increased mortality with left ventricular systolic dysfunction and heart failure in adults with myotonic
dystrophy type 1. Am. Heart J. 2010; 160:1137-41, 41.
328. Groh WJ, Groh MR, Saha C, et al. Electrocardiographic abnormalities and sudden death in myotonic dystrophy type 1. N. Engl. J. Med.
2008; 358:2688-97.
329. Lafort P, de TC, Eymard B, et al. Cardiac involvement in genetically confirmed facioscapulohumeral muscular dystrophy. Neurology. 1998;
51:1454-6.
330. Stevenson WG, Perloff JK, Weiss JN, et al. Facioscapulohumeral muscular dystrophy: evidence for selective, genetic electrophysiologic
cardiac involvement. J Am Coll Cardiol. 1990; 15:292-9.
331. Costa J, Lopes CM, Barsheshet A, et al. Combined assessment of sex- and mutation-specific information for risk stratification in type 1 long
QT syndrome. Heart rhythm. 2012; 9:892-8.
332. Bai R, Napolitano C, Bloise R, et al. Yield of genetic screening in inherited cardiac channelopathies: how to prioritize access to genetic
testing. Circ. Arrhythm. Electrophysiol. 2009; 2:6-15.
333. Gehi AK, Duong TD, Metz LD, et al. Risk stratification of individuals with the Brugada electrocardiogram: a meta-analysis. J. Cardiovasc.
Electrophysiol. 2006; 17:577-83.
334. Kim JA, Lopes CM, Moss AJ, et al. Trigger-specific risk factors and response to therapy in long QT syndrome type 2. Heart rhythm. 2010;
7:1797-805.
335. Migdalovich D, Moss AJ, Lopes CM, et al. Mutation and gender-specific risk in type 2 long QT syndrome: implications for risk stratification
for life-threatening cardiac events in patients with long QT syndrome. Heart rhythm. 2011; 8:1537-43.
336. Nannenberg EA, Sijbrands EJ, Dijksman LM, et al. Mortality of inherited arrhythmia syndromes: insight into their natural history. Circ.
Cardiovasc. Genet. 2012; 5:183-9.
337. Kimbrough J, Moss AJ, Zareba W, et al. Clinical implications for affected parents and siblings of probands with long-QT syndrome.
Circulation. 2001; 104:557-62.
338. Kaufman ES, McNitt S, Moss AJ, et al. Risk of death in the long QT syndrome when a sibling has died. Heart rhythm. 2008; 5:831-6.
339. Wedekind H, Burde D, Zumhagen S, et al. QT interval prolongation and risk for cardiac events in genotyped LQTS-index children. Eur. J
Pediatr. 2009; 168:1107-15.
340. Goldenberg I, Horr S, Moss AJ, et al. Risk for life-threatening cardiac events in patients with genotype-confirmed long-QT syndrome and
normal-range corrected QT intervals. J Am Coll Cardiol. 2011; 57:51-9.
341. Tester DJ, Will ML, Haglund CM, et al. Effect of clinical phenotype on yield of long QT syndrome genetic testing. J Am Coll Cardiol. 2006;
47:764-8.

341

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
342. Priori SG, Napolitano C, Memmi M, et al. Clinical and molecular characterization of patients with catecholaminergic polymorphic
ventricular tachycardia. Circulation. 2002; 106:69-74.
343. Probst V, Veltmann C, Eckardt L, et al. Long-term prognosis of patients diagnosed with Brugada syndrome: results from the FINGER
Brugada syndrome registry. Circulation. 2010; 121:635-43.
344. Moss AJ, Zareba W, Hall WJ, et al. Effectiveness and limitations of beta-blocker therapy in congenital long-QT syndrome. Circulation. 2000;
101:616-23.
345. Zareba W, Moss AJ, Daubert JP, et al. Implantable cardioverter defibrillator in high-risk long QT syndrome patients. J. Cardiovasc.
Electrophysiol. 2003; 14:337-41.
346. Monnig G, Kobe J, Loher A, et al. Implantable cardioverter-defibrillator therapy in patients with congenital long-QT syndrome: a long-term
follow-up. Heart rhythm. 2005; 2:497-504.
347. Hayashi M, Denjoy I, Extramiana F, et al. Incidence and risk factors of arrhythmic events in catecholaminergic polymorphic ventricular
tachycardia. Circulation. 2009; 119:2426-34.
348. Delise P, Allocca G, Marras E, et al. Risk stratification in individuals with the Brugada type 1 ECG pattern without previous cardiac arrest:
usefulness of a combined clinical and electrophysiologic approach. Eur. Heart J. 2011; 32:169-76.
349. Hiraoka M, Takagi M, Yokoyama Y, et al. Prognosis and risk stratification of young adults with Brugada syndrome. Journal of
electrocardiology. 2013; 46:279-83.
350. Garson A, Jr., Dick M, Fournier A, et al. The long QT syndrome in children. An international study of 287 patients. Circulation. 1993;
87:1866-72.
351. Hobbs JB, Peterson DR, Moss AJ, et al. Risk of aborted cardiac arrest or sudden cardiac death during adolescence in the long-QT syndrome.
Jama. 2006; 296:1249-54.
352. Priori SG, Schwartz PJ, Napolitano C, et al. Risk stratification in the long-QT syndrome. N. Engl. J. Med. 2003; 348:1866-74.
353. Jons C, Moss AJ, Goldenberg I, et al. Risk of fatal arrhythmic events in long QT syndrome patients after syncope. J Am Coll Cardiol. 2010;
55:783-8.
354. Barsheshet A, Goldenberg I, Uchi J, et al. Mutations in cytoplasmic loops of the KCNQ1 channel and the risk of life-threatening events:
implications for mutation-specific response to beta-blocker therapy in type 1 long-QT syndrome. Circulation. 2012; 125:1988-96.
355. Vincent GM, Schwartz PJ, Denjoy I, et al. High efficacy of beta-blockers in long-QT syndrome type 1: contribution of noncompliance and
QT-prolonging drugs to the occurrence of beta-blocker treatment "failures". Circulation. 2009; 119:215-21.
356. Abu-Zeitone A, Peterson DR, Polonsky B, et al. Efficacy of different beta-blockers in the treatment of long QT syndrome. J Am Coll Cardiol.
2014; 64:1352-8.
357. Goldenberg I, Bradley J, Moss A, et al. Beta-blocker efficacy in high-risk patients with the congenital long-QT syndrome types 1 and 2:
implications for patient management. J. Cardiovasc. Electrophysiol. 2010; 21:893-901.
358. Sauer AJ, Moss AJ, McNitt S, et al. Long QT syndrome in adults. J Am Coll Cardiol. 2007; 49:329-37.
359. Steinberg C, Padfield GJ, Al-Sabeq B, et al. Experience with bisoprolol in long-QT1 and long-QT2 syndrome. J Interv. Card Electrophysiol.
2016.
342

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
360. Villain E, Denjoy I, Lupoglazoff JM, et al. Low incidence of cardiac events with beta-blocking therapy in children with long QT syndrome.
Eur. Heart J. 2004; 25:1405-11.
361. Moltedo JM, Kim JJ, Friedman RA, et al. Use of a cardioselective beta-blocker for pediatric patients with prolonged QT syndrome. Pediatr.
Cardiol. 2011; 32:63-6.
362. Schwartz PJ, Priori SG, Cerrone M, et al. Left cardiac sympathetic denervation in the management of high-risk patients affected by the
long-QT syndrome. Circulation. 2004; 109:1826-33.
363. Bos JM, Bos KM, Johnson JN, et al. Left cardiac sympathetic denervation in long QT syndrome: analysis of therapeutic nonresponders. Circ.
Arrhythm. Electrophysiol. 2013; 6:705-11.
364. Schneider HE, Steinmetz M, Krause U, et al. Left cardiac sympathetic denervation for the management of life-threatening ventricular
tachyarrhythmias in young patients with catecholaminergic polymorphic ventricular tachycardia and long QT syndrome. Clin. Res. Cardiol.
2013; 102:33-42.
365. Collura CA, Johnson JN, Moir C, et al. Left cardiac sympathetic denervation for the treatment of long QT syndrome and catecholaminergic
polymorphic ventricular tachycardia using video-assisted thoracic surgery. Heart rhythm. 2009; 6:752-9.
366. Hofferberth SC, Cecchin F, Loberman D, et al. Left thoracoscopic sympathectomy for cardiac denervation in patients with life-threatening
ventricular arrhythmias. J Thorac. Cardiovasc. Surg. 2014; 147:404-9.
367. Chattha IS, Sy RW, Yee R, et al. Utility of the recovery electrocardiogram after exercise: a novel indicator for the diagnosis and genotyping
of long QT syndrome? Heart rhythm. 2010; 7:906-11.
368. Aziz PF, Wieand TS, Ganley J, et al. Genotype- and mutation site-specific QT adaptation during exercise, recovery, and postural changes in
children with long-QT syndrome. Circ Arrhythm Electrophysiol. 2011; 4:867-73.
369. Laksman ZW, Hamilton RM, Chockalingam P, et al. Mutation location effect on severity of phenotype during exercise testing in type 1 long-
QT syndrome: impact of transmembrane and C-loop location. Journal of cardiovascular electrophysiology. 2013; 24:1015-20.
370. Sy RW, van der Werf C, Chattha IS, et al. Derivation and validation of a simple exercise-based algorithm for prediction of genetic testing in
relatives of LQTS probands. Circulation. 2011; 124:2187-94.
371. Spazzolini C, Mullally J, Moss AJ, et al. Clinical implications for patients with long QT syndrome who experience a cardiac event during
infancy. J Am Coll Cardiol. 2009; 54:832-7.
372. Zhang C, Kutyifa V, Moss AJ, et al. Long-QT Syndrome and Therapy for Attention Deficit/Hyperactivity Disorder. Journal of cardiovascular
electrophysiology. 2015; 26:1039-44.
373. Choy AM, Lang CC, Chomsky DM, et al. Normalization of acquired QT prolongation in humans by intravenous potassium. Circulation. 1997;
96:2149-54.
374. Kannankeril P, Roden DM and Darbar D. Drug-induced long QT syndrome. Pharmacol. Rev. 2010; 62:760-81.
375. Roston TM, Vinocur JM, Maginot KR, et al. Catecholaminergic polymorphic ventricular tachycardia in children: analysis of therapeutic
strategies and outcomes from an international multicenter registry. Circ. Arrhythm. Electrophysiol. 2015; 8:633-42.
376. Wilde AA, Bhuiyan ZA, Crotti L, et al. Left cardiac sympathetic denervation for catecholaminergic polymorphic ventricular tachycardia. N.
Engl. J. Med. 2008; 358:2024-9.
343

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
377. Li J, Liu Y, Yang F, et al. Video-assisted thoracoscopic left cardiac sympathetic denervation: a reliable minimally invasive approach for
congenital long-QT syndrome. Ann. Thorac. Surg. 2008; 86:1955-8.
378. van der Werf C, Kannankeril PJ, Sacher F, et al. Flecainide therapy reduces exercise-induced ventricular arrhythmias in patients with
catecholaminergic polymorphic ventricular tachycardia. J Am Coll Cardiol. 2011; 57:2244-54.
379. Watanabe H, van der Werf C, Roses-Noguer F, et al. Effects of flecainide on exercise-induced ventricular arrhythmias and recurrences in
genotype-negative patients with catecholaminergic polymorphic ventricular tachycardia. Heart rhythm. 2013; 10:542-7.
380. Somani R, Krahn AD, Healey JS, et al. Procainamide infusion in the evaluation of unexplained cardiac arrest: from the Cardiac Arrest
Survivors with Preserved Ejection Fraction Registry (CASPER). Heart rhythm. 2014; 11:1047-54.
381. Mizusawa Y, Morita H, Adler A, et al. Prognostic significance of fever-induced Brugada syndrome. Heart rhythm. 2016; 13:1515-20.
382. Priori SG, Gasparini M, Napolitano C, et al. Risk stratification in Brugada syndrome: results of the PRELUDE (PRogrammed ELectrical
stimUlation preDictive valuE) registry. J Am Coll Cardiol. 2012; 59:37-45.
383. Casado-Arroyo R, Berne P, Rao JY, et al. Long-term trends in newly diagnosed Brugada syndrome: implications for risk stratification. J Am
Coll Cardiol. 2016; 68:614-23.
384. Belhassen B, Rahkovich M, Michowitz Y, et al. Management of Brugada Syndrome: Thirty-Three-Year Experience Using
Electrophysiologically Guided Therapy With Class 1A Antiarrhythmic Drugs. Circ Arrhythm Electrophysiol. 2015; 8:1393-402.
385. Nademanee K, Veerakul G, Chandanamattha P, et al. Prevention of ventricular fibrillation episodes in Brugada syndrome by catheter
ablation over the anterior right ventricular outflow tract epicardium. Circulation. 2011; 123:1270-9.
386. Sunsaneewitayakul B, Yao Y, Thamaree S, et al. Endocardial mapping and catheter ablation for ventricular fibrillation prevention in
Brugada syndrome. J. Cardiovasc. Electrophysiol. 2012; 23 Suppl 1:S10-S6.
387. Zhang P, Tung R, Zhang Z, et al. Characterization of the epicardial substrate for catheter ablation of Brugada syndrome. Heart rhythm.
2016.
388. Brugada J, Pappone C, Berruezo A, et al. Brugada syndrome phenotype elimination by epicardial substrate ablation. Circ. Arrhythm.
Electrophysiol. 2015; 8:1373-81.
389. McNamara DA, Goldberger JJ, Berendsen MA, et al. Implantable defibrillators versus medical therapy for cardiac channelopathies.
Cochrane Database Syst. Rev. 2015:CD011168.
390. Sieira J, Ciconte G, Conte G, et al. Asymptomatic Brugada syndrome: clinical characterization and long-term prognosis. Circ. Arrhythm.
Electrophysiol. 2015; 8:1144-50.
391. Konigstein M, Rosso R, Topaz G, et al. Drug-induced Brugada syndrome: Clinical characteristics and risk factors. Heart rhythm. 2016;
13:1083-7.
392. Sroubek J, Probst V, Mazzanti A, et al. Programmed ventricular stimulation for risk stratification in the brugada syndrome: a pooled
analysis. Circulation. 2016; 133:622-30.
393. Sieira J, Conte G, Ciconte G, et al. Clinical characterisation and long-term prognosis of women with Brugada syndrome. Heart (British
Cardiac Society). 2016; 102:452-8.

344

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
394. Priori SG, Napolitano C, Gasparini M, et al. Natural history of Brugada syndrome: insights for risk stratification and management.
Circulation. 2002; 105:1342-7.
395. Fauchier L, Isorni MA, Clementy N, et al. Prognostic value of programmed ventricular stimulation in Brugada syndrome according to clinical
presentation: an updated meta-analysis of worldwide published data. Int J Cardiol. 2013; 168:3027-9.
396. Rodriguez-Manero M, Sacher F, de AC, et al. Monomorphic ventricular tachycardia in patients with Brugada syndrome: A multicenter
retrospective study. Heart rhythm. 2016; 13:669-82.
397. Sacher F, Probst V, Maury P, et al. Outcome after implantation of a cardioverter-defibrillator in patients with Brugada syndrome: a
multicenter study-part 2. Circulation. 2013; 128:1739-47.
398. Rosso R, Kogan E, Belhassen B, et al. J-point elevation in survivors of primary ventricular fibrillation and matched control subjects:
incidence and clinical significance. J Am Coll Cardiol. 2008; 52:1231-8.
399. Conte G, Sieira J, Ciconte G, et al. Implantable cardioverter-defibrillator therapy in Brugada syndrome: a 20-year single-center experience. J
Am Coll Cardiol. 2015; 65:879-88.
400. Miyazaki S, Uchiyama T, Komatsu Y, et al. Long-term complications of implantable defibrillator therapy in Brugada syndrome. Am J Cardiol.
2013; 111:1448-51.
401. Takagi M, Sekiguchi Y, Yokoyama Y, et al. Long-term prognosis in patients with Brugada syndrome based on Class II indication for
implantable cardioverter-defibrillator in the HRS/EHRA/APHRS Expert Consensus Statement: multicenter study in Japan. Heart rhythm.
2014; 11:1716-20.
402. Haissaguerre M, Sacher F, Nogami A, et al. Characteristics of recurrent ventricular fibrillation associated with inferolateral early
repolarization role of drug therapy. J Am Coll Cardiol. 2009; 53:612-9.
403. Tikkanen JT, Anttonen O, Junttila MJ, et al. Long-term outcome associated with early repolarization on electrocardiography. N. Engl. J.
Med. 2009; 361:2529-37.
404. Sinner MF, Porthan K, Noseworthy PA, et al. A meta-analysis of genome-wide association studies of the electrocardiographic early
repolarization pattern. Heart rhythm. 2012; 9:1627-34.
405. Adhikarla C, Boga M, Wood AD, et al. Natural history of the electrocardiographic pattern of early repolarization in ambulatory patients.
Am. J. Cardiol. 2011; 108:1831-5.
406. Siebermair J, Sinner MF, Beckmann BM, et al. Early repolarization pattern is the strongest predictor of arrhythmia recurrence in patients
with idiopathic ventricular fibrillation: results from a single centre long-term follow-up over 20 years. Europace : European pacing,
arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular
electrophysiology of the European Society of Cardiology. 2016; 18:718-25.
407. Tikkanen JT, Wichmann V, Junttila MJ, et al. Association of early repolarization and sudden cardiac death during an acute coronary event.
Circ. Arrhythm. Electrophysiol. 2012; 5:714-8.
408. Junttila MJ, Tikkanen JT, Kentta T, et al. Early repolarization as a predictor of arrhythmic and nonarrhythmic cardiac events in middle-aged
subjects. Heart rhythm. 2014; 11:1701-6.
409. Gaita F, Giustetto C, Bianchi F, et al. Short QT syndrome: pharmacological treatment. J Am Coll Cardiol. 2004; 43:1494-9.
345

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
410. Gollob MH, Redpath CJ and Roberts JD. The short QT syndrome: proposed diagnostic criteria. J Am Coll Cardiol. 2011; 57:802-12.
411. Villafane J, Atallah J, Gollob MH, et al. Long-term follow-up of a pediatric cohort with short QT syndrome. J Am Coll Cardiol. 2013; 61:1183-
91.
412. Mazzanti A, Kanthan A, Monteforte N, et al. Novel insight into the natural history of short QT syndrome. J Am Coll Cardiol. 2014; 63:1300-
8.
413. Iribarren C, Round AD, Peng JA, et al. Short QT in a cohort of 1.7 million persons: prevalence, correlates, and prognosis. Ann. Noninvasive.
Electrocardiol. 2014; 19:490-500.
414. Guerrier K, Kwiatkowski D, Czosek RJ, et al. Short QT interval prevalence and clinical outcomes in a pediatric population. Circ. Arrhythm.
Electrophysiol. 2015; 8:1460-4.
415. Bun SS, Maury P, Giustetto C, et al. Electrical storm in short-QT syndrome successfully treated with Isoproterenol. J. Cardiovasc.
Electrophysiol. 2012; 23:1028-30.
416. Dhutia H, Malhotra A, Parpia S, et al. The prevalence and significance of a short QT interval in 18,825 low-risk individuals including
athletes. Br. J Sports Med. 2016; 50:124-9.
417. Ling Z, Liu Z, Su L, et al. Radiofrequency ablation versus antiarrhythmic medication for treatment of ventricular premature beats from the
right ventricular outflow tract: prospective randomized study. Circ. Arrhythm. Electrophysiol. 2014; 7:237-43.
418. Liao Z, Zhan X, Wu S, et al. Idiopathic ventricular arrhythmias originating from the pulmonary sinus cusp: prevalence,
electrocardiographic/electrophysiological characteristics, and catheter ablation. J Am Coll Cardiol. 2015; 66:2633-44.
419. Morady F, Kadish AH, DiCarlo L, et al. Long-term results of catheter ablation of idiopathic right ventricular tachycardia. Circulation. 1990;
82:2093-9.
420. Yamada T, Litovsky SH and Kay GN. The left ventricular ostium: an anatomic concept relevant to idiopathic ventricular arrhythmias. Circ.
Arrhythm. Electrophysiol. 2008; 1:396-404.
421. Yamada T, McElderry HT, Doppalapudi H, et al. Idiopathic ventricular arrhythmias originating from the left ventricular summit: anatomic
concepts relevant to ablation. Circ. Arrhythm. Electrophysiol. 2010; 3:616-23.
422. Mountantonakis SE, Frankel DS, Tschabrunn CM, et al. Ventricular arrhythmias from the coronary venous system: Prevalence, mapping,
and ablation. Heart rhythm. 2015; 12:1145-53.
423. Doppalapudi H, Yamada T, Ramaswamy K, et al. Idiopathic focal epicardial ventricular tachycardia originating from the crux of the heart.
Heart rhythm. 2009; 6:44-50.
424. Konstantinidou M, Koektuerk B, Wissner E, et al. Catheter ablation of right ventricular outflow tract tachycardia: a simplified remote-
controlled approach. Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac
pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2011; 13:696-700.
425. Ouyang F, Fotuhi P, Ho SY, et al. Repetitive monomorphic ventricular tachycardia originating from the aortic sinus cusp:
electrocardiographic characterization for guiding catheter ablation. J Am Coll Cardiol. 2002; 39:500-8.
426. Tada H, Ito S, Naito S, et al. Idiopathic ventricular arrhythmia arising from the mitral annulus: a distinct subgroup of idiopathic ventricular
arrhythmias. J Am Coll Cardiol. 2005; 45:877-86.
346

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
427. Tada H, Tadokoro K, Miyaji K, et al. Idiopathic ventricular arrhythmias arising from the pulmonary artery: prevalence, characteristics, and
topography of the arrhythmia origin. Heart rhythm. 2008; 5:419-26.
428. Tada H, Tadokoro K, Ito S, et al. Idiopathic ventricular arrhythmias originating from the tricuspid annulus: Prevalence, electrocardiographic
characteristics, and results of radiofrequency catheter ablation. Heart rhythm. 2007; 4:7-16.
429. Kamioka M, Mathew S, Lin T, et al. Electrophysiological and electrocardiographic predictors of ventricular arrhythmias originating from the
left ventricular outflow tract within and below the coronary sinus cusps. Clin. Res. Cardiol. 2015; 104:544-54.
430. Nagashima K, Choi EK, Lin KY, et al. Ventricular arrhythmias near the distal great cardiac vein: challenging arrhythmia for ablation. Circ.
Arrhythm. Electrophysiol. 2014; 7:906-12.
431. Yamada T, Maddox WR, McElderry HT, et al. Radiofrequency catheter ablation of idiopathic ventricular arrhythmias originating from
intramural foci in the left ventricular outflow tract: efficacy of sequential versus simultaneous unipolar catheter ablation. Circ. Arrhythm.
Electrophysiol. 2015; 8:344-52.
432. Hai JJ, Chahal AA, Friedman PA, et al. Electrophysiologic characteristics of ventricular arrhythmias arising from the aortic mitral continuity-
potential role of the conduction system. J. Cardiovasc. Electrophysiol. 2015; 26:158-63.
433. Yamada T, McElderry HT, Okada T, et al. Idiopathic left ventricular arrhythmias originating adjacent to the left aortic sinus of valsalva:
electrophysiological rationale for the surface electrocardiogram. J. Cardiovasc. Electrophysiol. 2010; 21:170-6.
434. Doppalapudi H, Yamada T, McElderry HT, et al. Ventricular tachycardia originating from the posterior papillary muscle in the left ventricle:
a distinct clinical syndrome. Circ. Arrhythm. Electrophysiol. 2008; 1:23-9.
435. Yamada T, Doppalapudi H, McElderry HT, et al. Electrocardiographic and electrophysiological characteristics in idiopathic ventricular
arrhythmias originating from the papillary muscles in the left ventricle: relevance for catheter ablation. Circ. Arrhythm. Electrophysiol.
2010; 3:324-31.
436. Yokokawa M, Good E, Desjardins B, et al. Predictors of successful catheter ablation of ventricular arrhythmias arising from the papillary
muscles. Heart rhythm. 2010; 7:1654-9.
437. Crawford T, Mueller G, Good E, et al. Ventricular arrhythmias originating from papillary muscles in the right ventricle. Heart rhythm. 2010;
7:725-30.
438. Ban JE, Lee HS, Lee DI, et al. Electrophysiological characteristics related to outcome after catheter ablation of idiopathic ventricular
arrhythmia originating from the papillary muscle in the left ventricle. Korean Circ. J. 2013; 43:811-8.
439. Nogami A, Naito S, Tada H, et al. Demonstration of diastolic and presystolic Purkinje potentials as critical potentials in a macroreentry
circuit of verapamil-sensitive idiopathic left ventricular tachycardia. J Am Coll Cardiol. 2000; 36:811-23.
440. Liu Y, Fang Z, Yang B, et al. Catheter ablation of fascicular ventricular tachycardia: long-term clinical outcomes and mechanisms of
recurrence. Circ. Arrhythm. Electrophysiol. 2015; 8:1443-51.
441. Lin D, Hsia HH, Gerstenfeld EP, et al. Idiopathic fascicular left ventricular tachycardia: linear ablation lesion strategy for noninducible or
nonsustained tachycardia. Heart rhythm. 2005; 2:934-9.
442. Hassaguerre M, Shoda M, Jais P, et al. Mapping and ablation of idiopathic ventricular fibrillation. Circulation. 2002; 106:962-7.

347

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
443. Noda T, Shimizu W, Taguchi A, et al. Malignant entity of idiopathic ventricular fibrillation and polymorphic ventricular tachycardia initiated
by premature extrasystoles originating from the right ventricular outflow tract. J Am Coll Cardiol. 2005; 46:1288-94.
444. Hassaguerre M, Shah DC, Jais P, et al. Role of Purkinje conducting system in triggering of idiopathic ventricular fibrillation. Lancet (London,
England). 2002; 359:677-8.
445. Van HH, Zado ES, Haqqani H, et al. Catheter ablation of ventricular fibrillation: importance of left ventricular outflow tract and papillary
muscle triggers. Heart rhythm. 2014; 11:566-73.
446. Sadek MM, Benhayon D, Sureddi R, et al. Idiopathic ventricular arrhythmias originating from the moderator band: Electrocardiographic
characteristics and treatment by catheter ablation. Heart rhythm. 2015; 12:67-75.
447. Tester DJ, Medeiros-Domingo A, Will ML, et al. Unexplained drownings and the cardiac channelopathies: a molecular autopsy series. Mayo
Clin. Proc. 2011; 86:941-7.
448. Tzimas I, Zingraf JC, Bajanowski T, et al. The role of known variants of KCNQ1, KCNH2, KCNE1, SCN5A, and NOS1AP in water-related deaths.
Int. J Legal Med. 2016.
449. Anderson JH, Tester DJ, Will ML, et al. Whole-exome molecular autopsy after exertion-related sudden unexplained death in the young.
Circ. Cardiovasc. Genet. 2016; 9:259-65.
450. Wang D, Shah KR, Um SY, et al. Cardiac channelopathy testing in 274 ethnically diverse sudden unexplained deaths. Forensic Sci. Int. 2014;
237:90-9.
451. Kumar S, Peters S, Thompson T, et al. Familial cardiological and targeted genetic evaluation: low yield in sudden unexplained death and
high yield in unexplained cardiac arrest syndromes. Heart rhythm. 2013; 10:1653-60.
452. Ban JE, Park HC, Park JS, et al. Electrocardiographic and electrophysiological characteristics of premature ventricular complexes associated
with left ventricular dysfunction in patients without structural heart disease. Europace : European pacing, arrhythmias, and cardiac
electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European
Society of Cardiology. 2013; 15:735-41.
453. Lee W, Tay A, Subbiah RN, et al. Impact of Implantable Cardioverter Defibrillators on Survival of Patients with Centrifugal Left Ventricular
Assist Devices. Pacing Clin. Electrophysiol. 2015; 38:925-33.
454. Carballeira Pol L, Deyell MW, Frankel DS, et al. Ventricular premature depolarization QRS duration as a new marker of risk for the
development of ventricular premature depolarization-induced cardiomyopathy. Heart rhythm. 2014; 11:299-306.
455. Deyell MW, Park KM, Han Y, et al. Predictors of recovery of left ventricular dysfunction after ablation of frequent ventricular premature
depolarizations. Heart rhythm. 2012; 9:1465-72.
456. Del Carpio Munoz F, Syed FF, Noheria A, et al. Characteristics of premature ventricular complexes as correlates of reduced left ventricular
systolic function: study of the burden, duration, coupling interval, morphology and site of origin of PVCs. J. Cardiovasc. Electrophysiol.
2011; 22:791-8.
457. Olgun H, Yokokawa M, Baman T, et al. The role of interpolation in PVC-induced cardiomyopathy. Heart rhythm. 2011; 8:1046-9.
458. Hasdemir C, Ulucan C, Yavuzgil O, et al. Tachycardia-induced cardiomyopathy in patients with idiopathic ventricular arrhythmias: the
incidence, clinical and electrophysiologic characteristics, and the predictors. J. Cardiovasc. Electrophysiol. 2011; 22:663-8.
348

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
459. Baman TS, Lange DC, Ilg KJ, et al. Relationship between burden of premature ventricular complexes and left ventricular function. Heart
rhythm. 2010; 7:865-9.
460. Kanei Y, Friedman M, Ogawa N, et al. Frequent premature ventricular complexes originating from the right ventricular outflow tract are
associated with left ventricular dysfunction. Ann. Noninvasive. Electrocardiol. 2008; 13:81-5.
461. Hamon D, Blaye-Felice MS, Bradfield JS, et al. A new combined parameter to predict premature ventricular complexes induced
cardiomyopathy: impact and recognition of epicardial origin. J. Cardiovasc. Electrophysiol. 2016; 27:709-17.
462. Bogun F, Crawford T, Reich S, et al. Radiofrequency ablation of frequent, idiopathic premature ventricular complexes: comparison with a
control group without intervention. Heart rhythm. 2007; 4:863-7.
463. Zhong L, Lee YH, Huang XM, et al. Relative efficacy of catheter ablation vs antiarrhythmic drugs in treating premature ventricular
contractions: a single-center retrospective study. Heart rhythm. 2014; 11:187-93.
464. Kawamura M, Badhwar N, Vedantham V, et al. Coupling interval dispersion and body mass index are independent predictors of idiopathic
premature ventricular complex-induced cardiomyopathy. J. Cardiovasc. Electrophysiol. 2014; 25:756-62.
465. Yokokawa M, Good E, Crawford T, et al. Recovery from left ventricular dysfunction after ablation of frequent premature ventricular
complexes. Heart rhythm. 2013; 10:172-5.
466. Jeejeebhoy FM, Zelop CM, Lipman S, et al. Cardiac Arrest in Pregnancy: A Scientific Statement From the American Heart Association.
Circulation. 2015; 132:1747-73.
467. Creanga AA, Berg CJ, Ko JY, et al. Maternal Mortality and Morbidity in the United States: Where Are We Now? Journal of Women's Health.
2014; 23:3-9.
468. Kampman MA, Balci A, Groen H, et al. Cardiac function and cardiac events 1-year postpartum in women with congenital heart disease. Am.
Heart J. 2015; 169:298-304.
469. Drenthen W, Boersma E, Balci A, et al. Predictors of pregnancy complications in women with congenital heart disease. Eur. Heart J. 2010;
31:2124-32.
470. Mhyre JM, Tsen LC, Einav S, et al. Cardiac arrest during hospitalization for delivery in the United States, 1998-2011. Anesthesiology. 2014;
120:810-8.
471. Siu SC, Sermer M, Colman JM, et al. Prospective multicenter study of pregnancy outcomes in women with heart disease. Circulation. 2001;
104:515-21.
472. Einav S, Kaufman N and Sela HY. Maternal cardiac arrest and perimortem caesarean delivery: evidence or expert-based? Resuscitation.
2012; 83:1191-200.
473. Citro R, Giudice R, Mirra M, et al. Is Tako-tsubo syndrome in the postpartum period a clinical entity different from peripartum
cardiomyopathy? J Cardiovasc Med (Hagerstown). 2013; 14:568-75.
474. Seth R, Moss AJ, McNitt S, et al. Long QT syndrome and pregnancy. J Am Coll Cardiol. 2007; 49:1092-8.
475. Katz V, Balderston K and DeFreest M. Perimortem cesarean delivery: were our assumptions correct? Am J Obstet. Gynecol. 2005;
192:1916-20.

349

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
476. Dijkman A, Huisman CM, Smit M, et al. Cardiac arrest in pregnancy: increasing use of perimortem caesarean section due to emergency
skills training? BJOG. 2010; 117:282-7.
477. Colletti PM, Lee KH and Elkayam U. Cardiovascular imaging of the pregnant patient. AJR. Am J Roentgenol. 2013; 200:515-21.
478. Natale A, Davidson T, Geiger MJ, et al. Implantable cardioverter-defibrillators and pregnancy: a safe combination? Circulation. 1997;
96:2808-12.
479. Damilakis J, Theocharopoulos N, Perisinakis K, et al. Conceptus radiation dose and risk from cardiac catheter ablation procedures.
Circulation. 2001; 104:893-7.
480. Preliminary report: effect of encainide and flecainide on mortality in a randomized trial of arrhythmia suppression after myocardial
infarction. The Cardiac Arrhythmia Suppression Trial (CAST) Investigators. N. Engl. J. Med. 1989; 321:406-12.
481. Effect of the antiarrhythmic agent moricizine on survival after myocardial infarction. The Cardiac Arrhythmia Suppression Trial II
Investigators. N. Engl. J. Med. 1992; 327:227-33.
482. Wyse DG, Friedman PL, Brodsky MA, et al. Life-threatening ventricular arrhythmias due to transient or correctable causes: high risk for
death in follow-up. J Am Coll Cardiol. 2001; 38:1718-24.
483. Monnig G, Kobe J, Loher A, et al. Role of implantable cardioverter defibrillator therapy in patients with acquired long QT syndrome: a long-
term follow-up. Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac pacing,
arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2012; 14:396-401.
484. Antman EM, Wenger TL, Butler VP, Jr., et al. Treatment of 150 cases of life-threatening digitalis intoxication with digoxin-specific Fab
antibody fragments. Final report of a multicenter study. Circulation. 1990; 81:1744-52.
485. Chan BS and Buckley NA. Digoxin-specific antibody fragments in the treatment of digoxin toxicity. Clin. Toxicol. . 2014; 52:824-36.
486. Hauptman PJ and Kelly RA. Digitalis. Circulation. 1999; 99:1265-70.
487. Kelly RA and Smith TW. Recognition and management of digitalis toxicity. Am. J. Cardiol. 1992; 69:108G-18G.
488. Osmonov D, Erdinler I, Ozcan KS, et al. Management of patients with drug-induced atrioventricular block. Pacing Clin. Electrophysiol. 2012;
35:804-10.
489. Tzivoni D, Banai S, Schuger C, et al. Treatment of torsade de pointes with magnesium sulfate. Circulation. 1988; 77:392-7.
490. Keren A, Tzivoni D, Gavish D, et al. Etiology, warning signs and therapy of torsade de pointes. A study of 10 patients. Circulation. 1981;
64:1167-74.
491. Yang T and Roden DM. Extracellular potassium modulation of drug block of IKr. Implications for torsade de pointes and reverse use-
dependence. Circulation. 1996; 93:407-11.
492. Hellestrand KJ, Burnett PJ, Milne JR, et al. Effect of the antiarrhythmic agent flecainide acetate on acute and chronic pacing thresholds.
Pacing Clin. Electrophysiol. 1983; 6:892-9.
493. Echt DS, Black JN, Barbey JT, et al. Evaluation of antiarrhythmic drugs on defibrillation energy requirements in dogs. Sodium channel block
and action potential prolongation. Circulation. 1989; 79:1106-17.
494. Crijns HJ, Van Gelder IC and Lie KI. Supraventricular tachycardia mimicking ventricular tachycardia during flecainide treatment. Am. J.
Cardiol. 1988; 62:1303-6.
350

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
495. Bajaj AK, Woosley RL and Roden DM. Acute electrophysiologic effects of sodium administration in dogs treated with O-desmethyl
encainide. Circulation. 1989; 80:994-1002.
496. Myerburg RJ, Kessler KM, Cox MM, et al. Reversal of proarrhythmic effects of flecainide acetate and encainide hydrochloride by
propranolol. Circulation. 1989; 80:1571-9.
497. Schwartz PJ and Woosley RL. Predicting the Unpredictable: Drug-Induced QT Prolongation and Torsades de Pointes. J Am Coll Cardiol.
2016; 67:1639-50.
498. Basso C, Burke M, Fornes P, et al. Guidelines for autopsy investigation of sudden cardiac death. Virchows Arch. 2008; 452:11-8.
499. Thorne SA, Barnes I, Cullinan P, et al. Amiodarone-associated thyroid dysfunction: risk factors in adults with congenital heart disease.
Circulation. 1999; 100:149-54.
500. Deal BJ, Scagliotti D, Miller SM, et al. Electrophysiologic drug testing in symptomatic ventricular arrhythmias after repair of tetralogy of
Fallot. Am J Cardiol. 1987; 59:1380-5.
501. Gatzoulis MA, Till JA, Somerville J, et al. Mechanoelectrical interaction in tetralogy of Fallot. QRS prolongation relates to right ventricular
size and predicts malignant ventricular arrhythmias and sudden death. Circulation. 1995; 92:231-7.
502. Koyak Z, Harris L, de Groot JR, et al. Sudden cardiac death in adult congenital heart disease. Circulation. 2012; 126:1944-54.
503. Diller GP, Kempny A, Liodakis E, et al. Left ventricular longitudinal function predicts life-threatening ventricular arrhythmia and death in
adults with repaired tetralogy of fallot. Circulation. 2012; 125:2440-6.
504. Harrison DA, Harris L, Siu SC, et al. Sustained ventricular tachycardia in adult patients late after repair of tetralogy of Fallot. J Am Coll
Cardiol. 1997; 30:1368-73.
505. Knauth AL, Gauvreau K, Powell AJ, et al. Ventricular size and function assessed by cardiac MRI predict major adverse clinical outcomes late
after tetralogy of Fallot repair. Heart (British Cardiac Society). 2008; 94:211-6.
506. Therrien J, Siu SC, Harris L, et al. Impact of pulmonary valve replacement on arrhythmia propensity late after repair of tetralogy of Fallot.
Circulation. 2001; 103:2489-94.
507. Therrien J, Provost Y, Merchant N, et al. Optimal timing for pulmonary valve replacement in adults after tetralogy of Fallot repair. Am J
Cardiol. 2005; 95:779-82.
508. Harrild DM, Berul CI, Cecchin F, et al. Pulmonary valve replacement in tetralogy of Fallot: impact on survival and ventricular tachycardia.
Circulation. 2009; 119:445-51.
509. Adamson L, Vohra HA and Haw MP. Does pulmonary valve replacement post repair of tetralogy of Fallot improve right ventricular
function? Interactive cardiovascular and thoracic surgery. 2009; 9:520-7.
510. Sabate Rotes A, Connolly HM, Warnes CA, et al. Ventricular arrhythmia risk stratification in patients with tetralogy of Fallot at the time of
pulmonary valve replacement. Circ Arrhythm Electrophysiol. 2015; 8:110-6.
511. Tsai SF, Chan DP, Ro PS, et al. Rate of inducible ventricular arrhythmia in adults with congenital heart disease. Am J Cardiol. 2010; 106:730-
6.
512. Garson A, Jr., Porter CB, Gillette PC, et al. Induction of ventricular tachycardia during electrophysiologic study after repair of tetralogy of
Fallot. J Am Coll Cardiol. 1983; 1:1493-502.
351

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
513. Chandar JS, Wolff GS, Garson A, Jr., et al. Ventricular arrhythmias in postoperative tetralogy of Fallot. Am J Cardiol. 1990; 65:655-61.
514. Kella DK, Merchant FM, Veledar E, et al. Lesion-specific differences for implantable cardioverter defibrillator therapies in adults with
congenital heart disease. Pacing Clin Electrophysiol. 2014; 37:1492-8.
515. Santharam S, Hudsmith L, Thorne S, et al. Long-term follow-up of implantable cardioverter-defibrillators in adult congenital heart disease
patients: indications and outcomes. Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working
groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2017; 19:407-13.
516. Vehmeijer JT, Brouwer TF, Limpens J, et al. Implantable cardioverter-defibrillators in adults with congenital heart disease: a systematic
review and meta-analysis. European heart journal. 2016; 37:1439-48.
517. Moore JP, Mondesert B, Lloyd MS, et al. Clinical Experience With the Subcutaneous Implantable Cardioverter-Defibrillator in Adults With
Congenital Heart Disease. Circ Arrhythm Electrophysiol. 2016; 9.
518. Okamura H, McLeod CJ, DeSimone CV, et al. Right Parasternal Lead Placement Increases Eligibility for Subcutaneous Implantable
Cardioverter Defibrillator Therapy in Adults With Congenital Heart Disease. Circulation journal : official journal of the Japanese Circulation
Society. 2016; 80:1328-35.
519. Yap SC, Roos-Hesselink JW, Hoendermis ES, et al. Outcome of implantable cardioverter defibrillators in adults with congenital heart
disease: a multi-centre study. European heart journal. 2007; 28:1854-61.
520. Khairy P, Landzberg MJ, Gatzoulis MA, et al. Value of programmed ventricular stimulation after tetralogy of fallot repair: a multicenter
study. Circulation. 2004; 109:1994-2000.
521. Khairy P, Harris L, Landzberg MJ, et al. Implantable cardioverter-defibrillators in tetralogy of Fallot. Circulation. 2008; 117:363-70.
522. Zeppenfeld K, Schalij MJ, Bartelings MM, et al. Catheter ablation of ventricular tachycardia after repair of congenital heart disease:
electroanatomic identification of the critical right ventricular isthmus. Circulation. 2007; 116:2241-52.
523. van Zyl M, Kapa S, Padmanabhan D, et al. Mechanism and outcomes of catheter ablation for ventricular tachycardia in adults with repaired
congenital heart disease. Heart rhythm. 2016; 13:1449-54.
524. Kapel GF, Reichlin T, Wijnmaalen AP, et al. Left-sided ablation of ventricular tachycardia in adults with repaired tetralogy of Fallot: a case
series. Circ Arrhythm Electrophysiol. 2014; 7:889-97.
525. Kapel GF, Reichlin T, Wijnmaalen AP, et al. Re-entry using anatomically determined isthmuses: a curable ventricular tachycardia in repaired
congenital heart disease. Circ. Arrhythm. Electrophysiol. 2015; 8:102-9.
526. Kapel GF, Sacher F, Dekkers OM, et al. Arrhythmogenic anatomical isthmuses identified by electroanatomical mapping are the substrate
for ventricular tachycardia in repaired tetralogy of Fallot. European heart journal. 2017; 38:268-76.
527. Khairy P, Harris L, Landzberg MJ, et al. Sudden death and defibrillators in transposition of the great arteries with intra-atrial baffles: a
multicenter study. Circ Arrhythm Electrophysiol. 2008; 1:250-7.
528. Tutarel O, Kempny A, Alonso-Gonzalez R, et al. Congenital heart disease beyond the age of 60: emergence of a new population with high
resource utilization, high morbidity, and high mortality. European heart journal. 2014; 35:725-32.

352

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
529. Koyak Z, de Groot JR, Bouma BJ, et al. Sudden cardiac death in adult congenital heart disease: can the unpredictable be foreseen?
Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac pacing, arrhythmias,
and cardiac cellular electrophysiology of the European Society of Cardiology. 2017; 19:401-6.
530. Engelfriet P, Boersma E, Oechslin E, et al. The spectrum of adult congenital heart disease in Europe: morbidity and mortality in a 5 year
follow-up period. The Euro Heart Survey on adult congenital heart disease. European heart journal. 2005; 26:2325-33.
531. Gallego P, Gonzalez AE, Sanchez-Recalde A, et al. Incidence and predictors of sudden cardiac arrest in adults with congenital heart defects
repaired before adult life. Am J Cardiol. 2012; 110:109-17.
532. Engelings CC, Helm PC, Abdul-Khaliq H, et al. Cause of death in adults with congenital heart disease - An analysis of the German National
Register for Congenital Heart Defects. Int J Cardiol. 2016; 211:31-6.
533. Fish FA, Gillette PC and Benson DW, Jr. Proarrhythmia, cardiac arrest and death in young patients receiving encainide and flecainide. The
Pediatric Electrophysiology Group. J Am Coll Cardiol. 1991; 18:356-65.
534. Stan MN, Sathananthan M, Warnes C, et al. Amiodarone-induced thyrotoxicosis in adults with congenital heart disease-clinical
presentation and response to therapy. Endocrine Practice. 2014; 21:33-40.
535. Silka MJ, Hardy BG, Menashe VD, et al. A population-based prospective evaluation of risk of sudden cardiac death after operation for
common congenital heart defects. J Am Coll Cardiol. 1998; 32:245-51.
536. Oechslin EN, Harrison DA, Connelly MS, et al. Mode of death in adults with congenital heart disease. Am J Cardiol. 2000; 86:1111-6.
537. Nieminen HP, Jokinen EV and Sairanen HI. Causes of late deaths after pediatric cardiac surgery: a population-based study. J Am Coll
Cardiol. 2007; 50:1263-71.
538. Verheugt CL, Uiterwaal CS, Grobbee DE, et al. Long-term prognosis of congenital heart defects: a systematic review. Int J Cardiol. 2008;
131:25-32.
539. Pillutla P, Shetty KD and Foster E. Mortality associated with adult congenital heart disease: Trends in the US population from 1979 to 2005.
Am Heart J. 2009; 158:874-9.
540. Verheugt CL, Uiterwaal CS, van der Velde ET, et al. Mortality in adult congenital heart disease. European heart journal. 2010; 31:1220-9.
541. Zomer AC, Vaartjes I, Uiterwaal CS, et al. Circumstances of death in adult congenital heart disease. Int J Cardiol. 2012; 154:168-72.
542. Diller GP, Kempny A, Alonso-Gonzalez R, et al. Survival Prospects and Circumstances of Death in Contemporary Adult Congenital Heart
Disease Patients Under Follow-Up at a Large Tertiary Centre. Circulation. 2015; 132:2118-25.
543. Raissadati A, Nieminen H, Haukka J, et al. Late Causes of Death After Pediatric Cardiac Surgery: A 60-Year Population-Based Study. J Am
Coll Cardiol. 2016; 68:487-98.
544. Teuwen CP, Ramdjan TT, Gotte M, et al. Non-sustained ventricular tachycardia in patients with congenital heart disease: An important
sign? Int J Cardiol. 2016; 206:158-63.
545. Wells R, Khairy P, Harris L, et al. Dofetilide for atrial arrhythmias in congenital heart disease: a multicenter study. Pacing Clin
Electrophysiol. 2009; 32:1313-8.
546. Afilalo J, Therrien J, Pilote L, et al. Geriatric congenital heart disease: burden of disease and predictors of mortality. J Am Coll Cardiol. 2011;
58:1509-15.
353

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
547. El Malti R, Liu H, Doray B, et al. A systematic variant screening in familial cases of congenital heart defects demonstrates the usefulness of
molecular genetics in this field. European journal of human genetics : EJHG. 2016; 24:228-36.
548. Abou Hassan OK, Fahed AC, Batrawi M, et al. NKX2-5 mutations in an inbred consanguineous population: genetic and phenotypic diversity.
Scientific reports. 2015; 5:8848.
549. Ellesoe SG, Johansen MM, Bjerre JV, et al. Familial Atrial Septal Defect and Sudden Cardiac Death: Identification of a Novel NKX2-5
Mutation and a Review of the Literature. Congenital heart disease. 2016; 11:283-90.
550. Cuypers JA, Opic P, Menting ME, et al. The unnatural history of an atrial septal defect: longitudinal 35 year follow up after surgical closure
at young age. Heart (British Cardiac Society). 2013; 99:1346-52.
551. Kuijpers JM, van der Bom T, van Riel AC, et al. Secundum atrial septal defect is associated with reduced survival in adult men. European
heart journal. 2015; 36:2079-86.
552. Khairy P, Aboulhosn J, Gurvitz MZ, et al. Arrhythmia burden in adults with surgically repaired tetralogy of Fallot: a multi-institutional study.
Circulation. 2010; 122:868-75.
553. Valente AM, Gauvreau K, Assenza GE, et al. Contemporary predictors of death and sustained ventricular tachycardia in patients with
repaired tetralogy of Fallot enrolled in the INDICATOR cohort. Heart (British Cardiac Society). 2014; 100:247-53.
554. Arya S, Kovach J, Singh H, et al. Arrhythmias and sudden death among older children and young adults following tetralogy of Fallot repair
in the current era: are previously reported risk factors still applicable? Congenital heart disease. 2014; 9:407-14.
555. Wu MH, Lu CW, Chen HC, et al. Arrhythmic burdens in patients with tetralogy of Fallot: a national database study. Heart rhythm. 2015;
12:604-9.
556. Drago F, Pazzano V, Di Mambro C, et al. Role of right ventricular three-dimensional electroanatomic voltage mapping for arrhythmic risk
stratification of patients with corrected tetralogy of Fallot or other congenital heart disease involving the right ventricular outflow tract. Int
J Cardiol. 2016; 222:422-9.
557. Kriebel T, Saul JP, Schneider H, et al. Noncontact mapping and radiofrequency catheter ablation of fast and hemodynamically unstable
ventricular tachycardia after surgical repair of tetralogy of Fallot. J Am Coll Cardiol. 2007; 50:2162-8.
558. Witte KK, Pepper CB, Cowan JC, et al. Implantable cardioverter-defibrillator therapy in adult patients with tetralogy of Fallot. Europace :
European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac
cellular electrophysiology of the European Society of Cardiology. 2008; 10:926-30.
559. Lange R, Horer J, Kostolny M, et al. Presence of a ventricular septal defect and the Mustard operation are risk factors for late mortality
after the atrial switch operation: thirty years of follow-up in 417 patients at a single center. Circulation. 2006; 114:1905-13.
560. Schwerzmann M, Salehian O, Harris L, et al. Ventricular arrhythmias and sudden death in adults after a Mustard operation for
transposition of the great arteries. European heart journal. 2009; 30:1873-9.
561. Wheeler M, Grigg L and Zentner D. Can we predict sudden cardiac death in long-term survivors of atrial switch surgery for transposition of
the great arteries? Congenital heart disease. 2014; 9:326-32.
562. Bouzeman A, Marijon E, de Guillebon M, et al. Implantable cardiac defibrillator among adults with transposition of the great arteries and
atrial switch operation: case series and review of literature. Int J Cardiol. 2014; 177:301-6.
354

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
563. Buber J, Ackley TJ, Daniels CJ, et al. Outcomes following the implantation of cardioverter-defibrillator for primary prevention in
transposition of the great arteries after intra-atrial baffle repair: a single-centre experience. Europace : European pacing, arrhythmias, and
cardiac electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the
European Society of Cardiology. 2016; 18:1016-22.
564. Backhoff D, Kerst G, Peters A, et al. Internal Cardioverter Defibrillator Indications and Therapies after Atrial Baffle Procedure for d-
Transposition of the Great Arteries: A Multicenter Analysis. Pacing Clin Electrophysiol. 2016; 39:1070-6.
565. Pundi KN, Pundi KN, Johnson JN, et al. Sudden cardiac death and late arrhythmias after the Fontan operation. Congenital heart disease.
2017; 12:17-23.
566. Sakamoto T, Nagashima M, Hiramatsu T, et al. Fontan circulation over 30 years. What should we learn from those patients? Asian
cardiovascular & thoracic annals. 2016; 24:765-71.
567. Alexander ME, Walsh EP, Saul JP, et al. Value of programmed ventricular stimulation in patients with congenital heart disease. Journal of
cardiovascular electrophysiology. 1999; 10:1033-44.
568. Silka MJ, Kron J, Dunnigan A, et al. Sudden cardiac death and the use of implantable cardioverter-defibrillators in pediatric patients. The
Pediatric Electrophysiology Society. Circulation. 1993; 87:800-7.
569. Berul CI, Van Hare GF, Kertesz NJ, et al. Results of a multicenter retrospective implantable cardioverter-defibrillator registry of pediatric
and congenital heart disease patients. J Am Coll Cardiol. 2008; 51:1685-91.
570. Khanna AD, Warnes CA, Phillips SD, et al. Single-center experience with implantable cardioverter-defibrillators in adults with complex
congenital heart disease. Am J Cardiol. 2011; 108:729-34.
571. Koyak Z, de Groot JR, Van Gelder IC, et al. Implantable cardioverter defibrillator therapy in adults with congenital heart disease: who is at
risk of shocks? Circ Arrhythm Electrophysiol. 2012; 5:101-10.
572. Khairy P, Van Hare GF, Balaji S, et al. PACES/HRS Expert Consensus Statement on the Recognition and Management of Arrhythmias in Adult
Congenital Heart Disease: developed in partnership between the Pediatric and Congenital Electrophysiology Society (PACES) and the Heart
Rhythm Society (HRS). Heart rhythm. 2014; 11:e102-65.
573. Bardy GH, Smith WM, Hood MA, et al. An entirely subcutaneous implantable cardioverter-defibrillator. N. Engl. J. Med. 2010; 363:36-44.
574. Olde Nordkamp LR, Dabiri AL, Boersma LV, et al. The entirely subcutaneous implantable cardioverter-defibrillator: initial clinical experience
in a large Dutch cohort. J Am Coll Cardiol. 2012; 60:1933-9.
575. Kbe J, Reinke F, Meyer C, et al. Implantation and follow-up of totally subcutaneous versus conventional implantable cardioverter-
defibrillators: a multicenter case-control study. Heart rhythm. 2013; 10:29-36.
576. de Bie MK, Thijssen J, van Rees JB, et al. Suitability for subcutaneous defibrillator implantation: results based on data from routine clinical
practice. Heart (British Cardiac Society). 2013; 99:1018-23.
577. Weiss R, Knight BP, Gold MR, et al. Safety and efficacy of a totally subcutaneous implantable-cardioverter defibrillator. Circulation. 2013;
128:944-53.
578. Olde Nordkamp LR, Warnaars JL, Kooiman KM, et al. Which patients are not suitable for a subcutaneous ICD: incidence and predictors of
failed QRS-T-wave morphology screening. J. Cardiovasc. Electrophysiol. 2014; 25:494-9.
355

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
579. Randles DA, Hawkins NM, Shaw M, et al. How many patients fulfil the surface electrocardiogram criteria for subcutaneous implantable
cardioverter-defibrillator implantation? Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working
groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2014; 16:1015-21.
580. Lambiase PD, Barr C, Theuns DA, et al. Worldwide experience with a totally subcutaneous implantable defibrillator: early results from the
EFFORTLESS S-ICD Registry. Eur. Heart J. 2014; 35:1657-65.
581. Groh WJ. Arrhythmias in the muscular dystrophies. Heart rhythm. 2012; 9:1890-5.
582. Burke MC, Gold MR, Knight BP, et al. Safety and efficacy of the totally subcutaneous implantable defibrillator: 2-year results from a pooled
analysis of the IDE study and EFFORTLESS registry. J Am Coll Cardiol. 2015; 65:1605-15.
583. Chung MK. The role of the wearable cardioverter defibrillator in clinical practice. Cardiol Clin. 2014; 32:253-70.
584. Chung MK, Szymkiewicz SJ, Shao M, et al. Aggregate national experience with the wearable cardioverter-defibrillator: event rates,
compliance, and survival. J Am Coll Cardiol. 2010; 56:194-203.
585. Klein HU, Meltendorf U, Reek S, et al. Bridging a temporary high risk of sudden arrhythmic death. Experience with the wearable
cardioverter defibrillator (WCD). Pacing Clin Electrophysiol. 2010; 33:353-67.
586. de Noronha SV, Behr ER, Papadakis M, et al. The importance of specialist cardiac histopathological examination in the investigation of
young sudden cardiac deaths. Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on
cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology. 2014; 16:899-907.
587. Wu Q, Zhang L, Zheng J, et al. Forensic Pathological Study of 1656 Cases of Sudden Cardiac Death in Southern China. Medicine (Baltimore).
2016; 95:e2707.
588. Vassalini M, Verzeletti A, Restori M, et al. An autopsy study of sudden cardiac death in persons aged 1-40 years in Brescia (Italy). J
Cardiovasc Med (Hagerstown). 2016; 17:446-53.
589. Tester DJ, Medeiros-Domingo A, Will ML, et al. Cardiac channel molecular autopsy: insights from 173 consecutive cases of autopsy-
negative sudden unexplained death referred for postmortem genetic testing. Mayo Clin. Proc. 2012; 87:524-39.
590. Tang Y, Stahl-Herz J and Sampson BA. Molecular diagnostics of cardiovascular diseases in sudden unexplained death. Cardiovasc Pathol.
2014; 23:1-4.
591. Papadakis M, Raju H, Behr ER, et al. Sudden cardiac death with autopsy findings of uncertain significance: potential for erroneous
interpretation. Circ. Arrhythm. Electrophysiol. 2013; 6:588-96.
592. Harmon KG, Drezner JA, Maleszewski JJ, et al. Pathogeneses of sudden cardiac death in national collegiate athletic association athletes.
Circ. Arrhythm. Electrophysiol. 2014; 7:198-204.
593. Bagnall RD, Das KJ, Duflou J, et al. Exome analysis-based molecular autopsy in cases of sudden unexplained death in the young. Heart
rhythm. 2014; 11:655-62.
594. Bagnall RD, Weintraub RG, Ingles J, et al. A prospective study of sudden cardiac death among children and young adults. N. Engl. J. Med.
2016; 374:2441-52.
595. Hill L, McIlfatrick S, Taylor B, et al. Patients' perception of implantable cardioverter defibrillator deactivation at the end of life. Palliat. Med.
2015; 29:310-23.
356

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
596. Kramer DB, Reynolds MR, Normand SL, et al. Hospice use following implantable cardioverter-defibrillator implantation in older patients:
results from the National Cardiovascular Data Registry. Circulation. 2016; 133:2030-7.
597. Buchhalter LC, Ottenberg AL, Webster TL, et al. Features and outcomes of patients who underwent cardiac device deactivation. JAMA
Intern. Med. 2014; 174:80-5.
598. Goldstein NE, Lampert R, Bradley E, et al. Management of implantable cardioverter defibrillators in end-of-life care. Ann. Intern. Med.
2004; 141:835-8.
599. Goldstein N, Carlson M, Livote E, et al. Brief communication: Management of implantable cardioverter-defibrillators in hospice: A
nationwide survey. Ann. Intern. Med. 2010; 152:296-9.
600. Berger JT, Gorski M and Cohen T. Advance health planning and treatment preferences among recipients of implantable cardioverter
defibrillators: an exploratory study. J Clin. Ethics. 2006; 17:72-8.
601. Dodson JA, Fried TR, Van Ness PH, et al. Patient preferences for deactivation of implantable cardioverter-defibrillators. JAMA Intern. Med.
2013; 173:377-9.
602. Goldstein NE, Mehta D, Siddiqui S, et al. "That's like an act of suicide" patients' attitudes toward deactivation of implantable defibrillators.
J Gen. Intern. Med. 2008; 23 Suppl 1:7-12.
603. Habal MV, Micevski V, Greenwood S, et al. How aware of advanced care directives are heart failure patients, and are they using them?
Can. J Cardiol. 2011; 27:376-81.
604. Kirkpatrick JN, Gottlieb M, Sehgal P, et al. Deactivation of implantable cardioverter defibrillators in terminal illness and end of life care.
Am. J. Cardiol. 2012; 109:91-4.
605. Kramer DB, Kesselheim AS, Salberg L, et al. Ethical and legal views regarding deactivation of cardiac implantable electrical devices in
patients with hypertrophic cardiomyopathy. Am. J. Cardiol. 2011; 107:1071-5.
606. Lewis KB, Stacey D and Matlock DD. Making decisions about implantable cardioverter-defibrillators from implantation to end of life: an
integrative review of patients' perspectives. Patient. 2014; 7:243-60.
607. Lewis KB, Nery PB and Birnie DH. Decision making at the time of ICD generator change: patients' perspectives. JAMA Intern. Med. 2014;
174:1508-11.
608. Hauptman PJ, Chibnall JT, Guild C, et al. Patient perceptions, physician communication, and the implantable cardioverter-defibrillator.
JAMA Intern. Med. 2013; 173:571-7.
609. Stewart GC, Weintraub JR, Pratibhu PP, et al. Patient expectations from implantable defibrillators to prevent death in heart failure. Journal
of cardiac failure. 2010; 16:106-13.
610. Ottenberg AL, Mueller PS, Topazian RJ, et al. "It's not broke, so let's not try to fix it": why patients decline a cardiovascular implantable
electronic device. Pacing Clin. Electrophysiol. 2014; 37:1306-14.
611. Yuhas J, Mattocks K, Gravelin L, et al. Patients' attitudes and perceptions of implantable cardioverter-defibrillators: potential barriers to
appropriate primary prophylaxis. Pacing Clin. Electrophysiol. 2012; 35:1179-87.

357

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society
612. Larsen G, Hallstrom A, McAnulty J, et al. Cost-effectiveness of the implantable cardioverter-defibrillator versus antiarrhythmic drugs in
survivors of serious ventricular tachyarrhythmias: results of the Antiarrhythmics Versus Implantable Defibrillators (AVID) economic analysis
substudy. Circulation. 2002; 105:2049-57.
613. O'Brien BJ, Connolly SJ, Goeree R, et al. Cost-effectiveness of the implantable cardioverter-defibrillator: results from the Canadian
Implantable Defibrillator Study (CIDS). Circulation. 2001; 103:1416-21.
614. Weiss JP, Saynina O, McDonald KM, et al. Effectiveness and cost-effectiveness of implantable cardioverter defibrillators in the treatment of
ventricular arrhythmias among medicare beneficiaries. Am J Med. 2002; 112:519-27.
615. Buxton M, Caine N, Chase D, et al. A review of the evidence on the effects and costs of implantable cardioverter defibrillator therapy in
different patient groups, and modelling of cost-effectiveness and cost-utility for these groups in a UK context. Health Technol. Assess.
2006; 10:iii-xi, 1.
616. Mark DB, Nelson CL, Anstrom KJ, et al. Cost-effectiveness of defibrillator therapy or amiodarone in chronic stable heart failure: results
from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT). Circulation. 2006; 114:135-42.
617. Zwanziger J, Hall WJ, Dick AW, et al. The cost effectiveness of implantable cardioverter-defibrillators: results from the Multicenter
Automatic Defibrillator Implantation Trial (MADIT)-II. J Am Coll Cardiol. 2006; 47:2310-8.
618. Mushlin AI, Hall WJ, Zwanziger J, et al. The cost-effectiveness of automatic implantable cardiac defibrillators: results from MADIT.
Multicenter Automatic Defibrillator Implantation Trial. Circulation. 1998; 97:2129-35.
619. Al-Khatib SM, Anstrom KJ, Eisenstein EL, et al. Clinical and economic implications of the Multicenter Automatic Defibrillator Implantation
Trial-II. Ann. Intern. Med. 2005; 142:593-600.
620. Sanders GD, Hlatky MA and Owens DK. Cost-effectiveness of implantable cardioverter-defibrillators. N. Engl. J. Med. 2005; 353:1471-80.
621. Smith T, Jordaens L, Theuns DA, et al. The cost-effectiveness of primary prophylactic implantable defibrillator therapy in patients with
ischaemic or non-ischaemic heart disease: a European analysis. Eur. Heart J. 2013; 34:211-9.
622. Cowie MR, Marshall D, Drummond M, et al. Lifetime cost-effectiveness of prophylactic implantation of a cardioverter defibrillator in
patients with reduced left ventricular systolic function: results of Markov modelling in a European population. Europace : European pacing,
arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular
electrophysiology of the European Society of Cardiology. 2009; 11:716-26.

358

2017 by the American College of Cardiology Foundation, American Heart Association, Inc., and Heart Rhythm Society

You might also like